Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis

Article Type
Changed
Fri, 04/26/2024 - 09:13
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

CT113004016_fig1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Sarcoptes%20scabiei%20mite%20(A)%2C%20ova%20(B)%2C%20and%20scybala%20(C)%20on%20microscopic%20evaluation.%3C%2Fp%3E

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

CT113004016_fig2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20%3Cem%3EPediculus%20humanus%3C%2Fem%3E%20(louse)%2C%20adult%20form.%3C%2Fp%3E

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

CT113004016_fig3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Maculae%20ceruleae%E2%80%94blue-grey%20macules%E2%80%94may%20be%20present%20on%20the%20skin%20secondary%20to%20%3Cem%3EPediculosis%3C%2Fem%3E%20infestation.%3C%2Fp%3E

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

CT113004016_fig4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Pediculosis%20nits%E2%80%94the%20egg%20cases%20of%20the%20parasite%E2%80%94may%20firmly%20attach%20to%20the%20hair%20shaft.%3C%2Fp%3E

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

CT113004016_fig5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20The%20pseudonit%20closely%20mimics%20pediculosis%20nits%20but%20consists%20of%20keratinized%20cell%20casts%20that%20are%20freely%20dislodged.%3C%2Fp%3E

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
Article PDF
Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Publications
Topics
Page Number
E16-E21
Sections
Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Author and Disclosure Information

Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.

The authors report no conflict of interest.

All images are in the public domain.

Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (alexis.grabow.strahan@live.mercer.edu).

Article PDF
Article PDF

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

CT113004016_fig1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Sarcoptes%20scabiei%20mite%20(A)%2C%20ova%20(B)%2C%20and%20scybala%20(C)%20on%20microscopic%20evaluation.%3C%2Fp%3E

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

CT113004016_fig2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20%3Cem%3EPediculus%20humanus%3C%2Fem%3E%20(louse)%2C%20adult%20form.%3C%2Fp%3E

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

CT113004016_fig3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Maculae%20ceruleae%E2%80%94blue-grey%20macules%E2%80%94may%20be%20present%20on%20the%20skin%20secondary%20to%20%3Cem%3EPediculosis%3C%2Fem%3E%20infestation.%3C%2Fp%3E

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

CT113004016_fig4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Pediculosis%20nits%E2%80%94the%20egg%20cases%20of%20the%20parasite%E2%80%94may%20firmly%20attach%20to%20the%20hair%20shaft.%3C%2Fp%3E

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

CT113004016_fig5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20The%20pseudonit%20closely%20mimics%20pediculosis%20nits%20but%20consists%20of%20keratinized%20cell%20casts%20that%20are%20freely%20dislodged.%3C%2Fp%3E

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.3 In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.4

Understanding how to care for this population begins with an accurate understanding of the term refugee. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.5,6

The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature7,8 and varies from inflammatory disorders to infectious and parasitic diseases.9 In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.10

When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.11 Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.8

Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.12 The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes.

Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.13 This review will focus on infestations by the scabies mite (Sarcoptes scabiei var hominis) and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings.

Scabies

Scabies is a parasitic skin infestation caused by the 8-legged mite Sarcoptes scabiei var hominis. The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.14,15 Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.16 Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature.

CT113004016_fig1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Sarcoptes%20scabiei%20mite%20(A)%2C%20ova%20(B)%2C%20and%20scybala%20(C)%20on%20microscopic%20evaluation.%3C%2Fp%3E

 

 

The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.17 In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.18 Scabies is more prevalent in children, with increased potential for secondary bacterial infections with Streptococcus and Staphylococcus species due to excoriation in unsanitary conditions. Secondary infection with Streptococcus pyogenes can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.19 However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential.

Presentation—The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.20 The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.19 Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.21

Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.22

Diagnosis—The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.23 A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy.

Treatment—First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the World Health Organization Model List of Essential Medications and are widely available for treating providers, even in resource-limited settings.24

Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.25Sarna is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours.

Pediculosis

Pediculosis is an infestation caused by the ectoparasite Pediculus humanus, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).26 Of the lice species, 2 require humans as hosts; one is P humanus and the other is Pthirus pubis (pubic lice). Pediculus humanus may be further classified into morphologies based largely on the affected area: body (P humanus corporis) or head (P humanus capitis), both of which will be discussed.27

CT113004016_fig2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20%3Cem%3EPediculus%20humanus%3C%2Fem%3E%20(louse)%2C%20adult%20form.%3C%2Fp%3E

 

 

Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.28 Transmission of P humanus capitis primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; P humanus corporis also may be transmitted via direct contact with affected individuals or clothing.

Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.29 Body lice are associated more often with domestic turbulence and displaced populations30 in comparison to head lice, which have broad demographic variables, most often affecting females and children.28 Fatty acids in adult male sebum make the scalp less hospitable to lice.

Presentation—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed maculae ceruleae (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.31 Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.32

CT113004016_fig3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Maculae%20ceruleae%E2%80%94blue-grey%20macules%E2%80%94may%20be%20present%20on%20the%20skin%20secondary%20to%20%3Cem%3EPediculosis%3C%2Fem%3E%20infestation.%3C%2Fp%3E

Diagnosis—The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).31

CT113004016_fig4.jpg
%3Cp%3E%3Cstrong%3EFIGURE%204.%3C%2Fstrong%3E%20Pediculosis%20nits%E2%80%94the%20egg%20cases%20of%20the%20parasite%E2%80%94may%20firmly%20attach%20to%20the%20hair%20shaft.%3C%2Fp%3E

Treatment—Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.33 The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.34,35 Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.22 Topical corticosteroids or emollients may be employed for residual pruritus.

CT113004016_fig5.jpg
%3Cp%3E%3Cstrong%3EFIGURE%205.%3C%2Fstrong%3E%20The%20pseudonit%20closely%20mimics%20pediculosis%20nits%20but%20consists%20of%20keratinized%20cell%20casts%20that%20are%20freely%20dislodged.%3C%2Fp%3E

Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.36 It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.37

Pediculosis corporis may transmit infectious agents including Bartonella quintana, (trench fever, endocarditis, bacillary angiomatosis), Borrelia recurrentis (louse-borne relapsing fever), and Rickettsia prowazekii (epidemic typhus).31,38,39 Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.40 It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.41

 

 

Future Considerations

Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.42

Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.43

Conclusion

Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.8,11,44 Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.45 Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.46 Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.

References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
References
  1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021
  2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html
  3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts
  4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/
  5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions
  6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees
  7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). Int J Dermatol. 2019;58:1341-1349. doi:10.1111/ijd.14619
  8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. Dermatol Clin. 2021;39:101-115. doi:10.1016/j.det.2020.08.010
  9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. J Eur Acad Dermatol Venereol. 2020;34:419-425. doi:10.1111/jdv.15909
  10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. J Infect Public Health. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012
  11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.J Travel Med. 2018;25. doi:10.1093/jtm/tay067
  12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. Trop Med Infect Dis. 2018;4. doi:10.3390/tropicalmed4010004
  13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases
  14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. Parasit Vectors. 2017;10:297. doi:10.1186/s13071-017-2234-1
  15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. J Am Acad Dermatol. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4
  16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. Dermatology. 2019;235:79-90. doi:10.1159/000495290
  17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. Lancet Infect Dis. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8
  18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. Lancet Infect Dis. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2
  19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. J Am Acad Dermatol. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109
  20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. Br Med J. 1942;2:1-4. doi:10.1136/bmj.2.4252.1
  21. Walton SF. The immunology of susceptibility and resistance to scabies. Parasit Immunol. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x
  22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. J Am Acad Dermatol. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110
  23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. PLoS Negl Trop Dis. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549
  24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02
  25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. J Eur Acad Dermatol Venereol. 2017;31:1248-1253. doi:10.1111/jdv.14351
  26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. Clin Microbiol Infect. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x
  27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. J Med Entomol. 2002;39:662-666. doi:10.1603/0022-2585-39.4.662
  28. Chosidow O. Scabies and pediculosis. Lancet. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1
  29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). Br J Dermatol. 2016;174:104-112. doi:10.1111/bjd.14226
  30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. Annu Rev Entomol. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739
  31. Ko CJ, Elston DM. Pediculosis. J Am Acad Dermatol. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4
  32. Bloomfield D. Head lice. Pediatr Rev. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34
  33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). Fitzpatrick’s Dermatology in General Medicine. McGraw Hill; 2008:2029.
  34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. J Infect Dis. 2006;193:474-476. doi:10.1086/499279
  35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. JAMA Dermatol. 2014;150:273-279. doi:10.1001/jamadermatol.2013.6398
  36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html
  37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. Pediatrics. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746
  38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin Infect Dis. 2000;31:131-135. doi:10.1086/313890
  39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. Emerg Infect Dis. 2014;20:907-908. doi:10.3201/eid2005.131242
  40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. JAMA Dermatol. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818
  41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. J Emergency Med. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030
  42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis
  43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. Br J Dermatology. 2023;188:553-554. doi:10.1093/bjd/ljad001
  44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. Int J Dermatol. 2020;59:1299-1311. doi:10.1111/ijd.15063
  45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. Expert Rev Anti-infective Therapy. 2020;18:127-143. doi:10.1080/14787210.2020.1713099
  46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization
Page Number
E16-E21
Page Number
E16-E21
Publications
Publications
Topics
Article Type
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis
Display Headline
Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Strahan</fileName> <TBEID>0C02F581.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F581</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Clinical Review</storyname> <articleType>1</articleType> <TBLocation>Published-CT</TBLocation> <QCDate/> <firstPublished>20240425T131253</firstPublished> <LastPublished>20240425T132642</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240425T131253</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Strahan</byline> <bylineText>Alexis G. Strahan, MD, MSN; Dirk M. Elston, MD</bylineText> <bylineFull>Strahan</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E16-E21</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.1,2 The U</metaDescription> <articlePDF>301172</articlePDF> <teaserImage/> <title>Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>2</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2293</CMSID> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>infectious disease</keyword> <keyword> scabies</keyword> <keyword> pediculosis</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Original Article | 2293<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">234</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800270f.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Dermatologic Care for Refugees: Effective Management of Scabies and Pediculosis</title> <deck/> </itemMeta> <itemContent> <p class="abstract">There is a large burden of treatable dermatologic conditions in refugee populations. Parasitic infestations are particularly common when there are barriers to basic hygiene, crowded living or travel conditions, and lack of access to health care. Body lice are associated with anemia and can transmit a variety of diseases; chronic impetigo secondary to scabies is a leading cause of chronic kidney disease globally. Dermatologists have unique skills to identify skin infections, inflammatory diseases, and infestations. Appropriate dermatologic care has the potential to improve overall outcomes.</p> <p>Approximately 108 million individuals have been forcibly displaced across the globe as of 2022, 35 million of whom are formally designated as refugees.<sup>1,2</sup> The United States has coordinated resettlement of more refugee populations than any other country; the most common countries of origin are the Democratic Republic of the Congo, Syria, Afghanistan, and Myanmar.<sup>3</sup> In 2021, policy to increase the number of refugees resettled in the United States by more than 700% (from 15,000 up to 125,000) was established; since enactment, the United States has seen more than double the refugee arrivals in 2023 than the prior year, making medical care for this population increasingly relevant for the dermatologist.<sup>4</sup> </p> <p>Understanding how to care for this population begins with an accurate understanding of the term <i>refugee</i>. The United Nations defines a refugee as a person who is unwilling or unable to return to their country of nationality because of persecution or well-founded fear of persecution due to race, religion, nationality, membership in a particular social group, or political opinion. This term grants a protected status under international law and encompasses access to travel assistance, housing, cultural orientation, and medical evaluation upon resettlement.<sup>5,6</sup> <br/><br/>The burden of treatable dermatologic conditions in refugee populations ranges from 19% to 96% in the literature<sup>7,8</sup> and varies from inflammatory disorders to infectious and parasitic diseases.<sup>9</sup> In one study of 6899 displaced individuals in Greece, the prevalence of dermatologic conditions was higher than traumatic injury, cardiac disease, psychological conditions, and dental disease.<sup>10</sup> <br/><br/>When outlining differential diagnoses for parasitic infestations of the skin that affect refugee populations, helpful considerations include the individual’s country of origin, route traveled, and method of travel.<sup>11</sup> Parasitic infestations specifically are more common in refugee populations when there are barriers to basic hygiene, crowded living or travel conditions, or lack of access to health care, which they may experience at any point in their home country, during travel, or in resettlement housing.<sup>8</sup> <br/><br/>Even with limited examination and diagnostic resources, the skin is the most accessible first indication of patients’ overall well-being and often provides simple diagnostic clues—in combination with contextualization of the patient’s unique circumstances—necessary for successful diagnosis and treatment of scabies and pediculosis.<sup>12</sup> The dermatologist working with refugee populations may be the first set of eyes available and trained to discern skin infestations and therefore has the potential to improve overall outcomes. <br/><br/>Some parasitic infestations in refugee populations may fall under the category of neglected tropical diseases, including scabies, ascariasis, trypanosomiasis, leishmaniasis, and schistosomiasis; they affect an estimated 1 billion individuals across the globe but historically have been underrepresented in the literature and in health policy due in part to limited access to care.<sup>13</sup> This review will focus on infestations by the scabies mite (<i>Sarcoptes scabiei </i>var <i>hominis</i>)<i> </i>and the human louse, as these frequently are encountered, easily diagnosed, and treatable by trained clinicians, even in resource-limited settings. </p> <h3>Scabies </h3> <p>Scabies is a parasitic skin infestation caused by the 8-legged mite <i>Sarcoptes scabiei </i>var<i> hominis.</i> The female mite begins the infestation process via penetration of the epidermis, particularly the stratum corneum, and commences laying eggs (Figure 1). The subsequent larvae emerge 48 to 72 hours later and remain burrowed in the epidermis. The larvae mature over the next 10 to 14 days and continue the reproductive cycle.<sup>14,15</sup> Symptoms of infestation occurs due to a hypersensitivity reaction to the mite and its by-products.<sup>16</sup> Transmission of the mite primarily occurs via direct (skin-to-skin) contact with infected individuals or environmental surfaces for 24 to36 hours in specific conditions, though the latter source has been debated in the literature. </p> <p>The method of transmission is particularly important when considering care for refugee populations. Scabies is found most often in those living in or traveling from tropical regions including East Asia, Southeast Asia, Oceania, and Latin America.<sup>17</sup> In displaced or refugee populations, a lack of access to basic hygiene, extended travel in close quarters, and suboptimal health care access all may lead to an increased incidence of untreated scabies infestations.<sup>18</sup> Scabies is more prevalent in children, with increased potential for secondary bacterial infections with <i>Streptococcus</i> and <i>Staphylococcus</i> species due to excoriation in unsanitary conditions. Secondary infection with <i>Streptococcus pyogenes</i> can lead to acute poststreptococcal glomerulonephritis, which accounts for a large burden of chronic kidney disease in affected populations.<sup>19</sup> However, scabies may be found in any population, regardless of hygiene or health care access. Treating health care providers should keep a broad differential. <br/><br/><i>Presentation—</i>The latency of scabies symptoms is 2 to 6 weeks in a primary outbreak and may be as short as 1 to 3 days with re-infestation, following the course of delayed-type hypersensitivity.<sup>20</sup> The initial hallmark symptom is pruritus with increased severity in the evening. Visible lesions, excoriations, and burrows associated with scattered vesicles or pustules may be seen over the web spaces of the hands and feet, volar surfaces of the wrists, axillae, waist, genitalia, inner thighs, or buttocks.<sup>19</sup> Chronic infestation often manifests with genital nodules. In populations with limited access to health care, there are reports of a sensitization phenomenon in which the individual may become less symptomatic after 4 to 6 weeks and yet be a potential carrier of the mite.<sup>21</sup><i> <br/><br/></i>Those with compromised immune function, such as individuals living with HIV or severe malnutrition, may present with crusted scabies, a variant that manifests as widespread hyperkeratotic scaling with more pronounced involvement of the head, neck, and acral areas. In contrast to classic scabies, crusted scabies is associated with minimal pruritus.<sup>22</sup> <br/><br/><i>Diagnosis—</i>The diagnosis of scabies is largely clinical with confirmation through skin scrapings. The International Alliance for Control of Scabies has established diagnostic criteria that include a combination of clinical findings, history, and visualization of mites.<sup>23</sup> A dermatologist working with refugee populations may employ any combination of history (eg, nocturnal itch, exposure to an affected individual) or clinical findings along with a high degree of suspicion in those with elevated risk. Visualization of mites is helpful to confirm the diagnosis and may be completed with the application of mineral oil at the terminal end of a burrow, skin scraping with a surgical blade or needle, and examination under light microscopy. <br/><br/><i>Treatment—</i>First-line treatment for scabies consists of application of permethrin cream 5% on the skin of the neck to the soles of the feet, which is to be left on for 8 to 14 hours followed by rinsing. Re-application is recommended in 1 to 2 weeks. Oral ivermectin is a reasonable alternative to permethrin cream due to its low cost and easy administration in large affected groups. It is not labeled for use in pregnant women or children weighing less than 15 kg but has no selective fetal toxicity. Treatment of scabies with ivermectin has the benefit of treating many other parasitic infections. Both medications are on the <i>World Health Organization Model List of Essential Medications</i> and are widely available for treating providers, even in resource-limited settings.<sup>24</sup> <br/><br/>Much of the world still uses benzyl benzoate or precipitated sulfur ointment to treat scabies, and some botanicals used in folk medicine have genuine antiscabetic properties. Pruritus may persist for 1 to 4 weeks following treatment and does not indicate treatment failure. Topical camphor and menthol preparations, low-potency topical corticosteroids, or emollients all may be employed for relief.<sup>25</sup> <em>Sarna</em> is a Spanish term for scabies and has become the proprietary name for topical antipruritic agents. Additional methods of treatment and prevention include washing clothes and linens in hot water and drying on high heat. If machine washing is not available, clothing and linens may be sealed in a plastic bag for 72 hours. </p> <h3>Pediculosis </h3> <p>Pediculosis is an infestation caused by the ectoparasite <i>Pediculus humanus</i>, an obligate, sesame seed–sized louse that feeds exclusively on the blood of its host (Figure 2).<sup>26</sup> Of the lice species, 2 require humans as hosts; one is <i>P humanus</i> and the other is <i>Pthirus pubis</i> (pubic lice). <i>Pediculus humanus </i>may be further classified into morphologies based largely on the affected area: body (<i>P humanus</i> <i>corporis)</i> or head (<i>P humanus</i> <i>capitis)</i>, both of which will be discussed.<sup>27</sup> </p> <p>Lice primarily attach to clothing and hair shafts, then transfer to the skin for blood feeds. Females lay eggs that hatch 6 to 10 days later, subsequently maturing into adults. The lifespan of these parasites with regular access to a host is 1 to 3 months for head lice and 18 days for body lice vs only 3 to 5 days without a host.<sup>28</sup> Transmission of <i>P humanus capitis </i>primarily occurs via direct contact with affected individuals, either head-to-head contact or sharing of items such as brushes and headscarves; <i>P humanus corporis</i> also may be transmitted via direct contact with affected individuals or clothing. <br/><br/>Pediculosis is an important infestation to consider when providing care for refugee populations. Risk factors include lack of access to basic hygiene, including regular bathing or laundering of clothing, and crowded conditions that make direct person-to-person contact with affected individuals more likely.<sup>29</sup> Body lice are associated more often with domestic turbulence and displaced populations<sup>30</sup> in comparison to head lice, which have broad demographic variables, most often affecting females and children.<sup>28</sup> Fatty acids in adult male sebum make the scalp less hospitable to lice. <br/><br/><i>Presentation</i>—The most common clinical manifestation of pediculosis is pruritus. Cutaneous findings can include papules, wheals, or hemorrhagic puncta secondary to the louse bite. Due to the Tyndall effect of deep hemosiderin pigment, blue-grey macules termed <i>maculae ceruleae</i> (Figure 3) also may be present in chronic infestations of pediculosis pubis, in contrast to pediculosis capitis or corporis.<sup>31</sup> Body louse infestation is associated with a general pruritus concentrated on the neck, shoulders, and waist—areas where clothing makes the most direct contact. Lesions may be visible and include eczematous patches with excoriation and possible secondary bacterial infection. Chronic infestation may exhibit lichenification or hyperpigmentation in associated areas. Head lice most often manifest with localized scalp pruritus and associated excoriation and cervical or occipital lymphadenopathy.<sup>32</sup> <br/><br/><i>Diagnosis—</i>The diagnosis of pediculosis is clinical, with confirmation requiring direct examination of the insect or nits (the egg case of the parasite)(Figure 4). Body lice and associated nits can be visualized on clothing seams near areas of highest body temperature, particularly the waistband. Head lice may be visualized crawling on hair shafts or on a louse comb. Nits are firmly attached to hair shafts and are visible to the naked eye, whereas pseudonits slide freely along the hair shaft and are not a manifestation of louse infestation (Figure 5).<sup>31</sup> <br/><br/><i>Treatment—</i>Treatment varies by affected area. Pediculosis corporis may be treated with permethrin cream 5% applied to the entire body and left on for 8 to 10 hours, but this may not be necessary if facilities are available to wash and dry clothing.<sup>33</sup> The use of oral ivermectin and permethrin-impregnated underwear both have been proposed.<sup>34,35</sup> Treatment of pediculosis capitis may be accomplished with a variety of topical pediculicides including permethrin, pyrethrum with piperonyl butoxide, dimethicone, malathion, benzyl alcohol, spinosad, and topical ivermectin.<sup>22</sup> Topical corticosteroids or emollients may be employed for residual pruritus. <br/><br/>Equally important is environmental elimination of infestation. Clothing should be discarded if possible or washed and dried using high heat. If neither approach is possible or appropriate, clothing may be sealed in a plastic bag for 2 weeks or treated with a pediculicide. Nit combing is an important adjunct in the treatment of pediculosis capitis.<sup>36</sup> It is important to encourage return to work and/or school immediately after treatment. “No nit” policies are more harmful to education than helpful for prevention of investation.<sup>37<br/><br/></sup>Pediculosis corporis may transmit infectious agents including <i>Bartonella quintana</i>, (trench fever, endocarditis, bacillary angiomatosis), <i>Borrelia recurrentis</i> (louse-borne relapsing fever), and <i>Rickettsia prowazekii</i> (epidemic typhus).<sup>31,38,39</sup> Additionally, severe pediculosis infestations have the potential to cause chronic blood loss in affected populations. In a study of patients with active pediculosis infestation, mean hemoglobin values were found to be 2.5 g/dL lower than a matched population without infestation.<sup>40</sup> It is important to consider pediculosis as a risk for iron-deficiency anemia in populations who are known to lack access to regular medical evaluation.<sup>41</sup> </p> <h3>Future Considerations </h3> <p>Increased access to tools and education for clinicians treating refugee populations is key to reducing the burden of parasitic skin disease and related morbidity and mortality in vulnerable groups both domestically and globally. One such tool, the Skin NTDs App, was launched by the World Health Organization in 2020. It is available for free for Android and iOS devices to assist clinicians in the field with the diagnosis and treatment of neglected tropical diseases—including scabies—that may affect refugee populations.<sup>42</sup></p> <p>Additionally, to both improve access and limit preventable sequelae, future investigations into appropriate models of community-based care are paramount. The model of community-based care is centered on the idea of care provision that prioritizes safety, accessibility, affordability, and acceptability in an environment closest to vulnerable populations. The largest dermatologic society, the International League of Dermatological Societies, formed a Migrant Health Dermatology Working Group that prioritizes understanding and improving care for refugee and migrant populations; this group hosted a summit in 2022, bringing together international subject matter leaders to discuss such models of care and set goals for the creation of tool kits for patients, frontline health care workers, and dermatologists.<sup>43</sup></p> <h3>Conclusion</h3> <p>Improvement in dermatologic care of refugee populations includes provision of culturally and linguistically appropriate care by trained clinicians, adequate access to the most essential medications, and basic physical or legal access to health care systems in general.<sup>8,11,44</sup> Parasitic infestations have the potential to remain asymptomatic for extended periods of time and result in spread to potentially nonendemic regions of resettlement.<sup>45</sup> Additionally, the psychosocial well-being of refugee populations upon resettlement may be negatively affected by stigma of disease processes such as scabies and pediculosis, leading to additional barriers to successful re-entry into the patient’s new environment.<sup>46</sup> Therefore, proper screening, diagnosis, and treatment of the most common parasitic infestations in this population have great potential to improve outcomes for large groups across the globe.</p> <h2>References</h2> <p class="reference"> 1. Monin K, Batalova J, Lai T. Refugees and Asylees in the United States. Migration Information Source. Published May 13, 2021. Accessed April 4, 2024. https://www.migrationpolicy.org/article/refugees-and-asylees-united-states-2021<br/><br/> 2. UNHCR. Figures at a Glance. UNHCR USA. Update June 14, 2023. Accessed April 4, 2024. https://www.unhcr.org/en-us/figures-at-a-glance.html<br/><br/> 3. UNHCR. Refugee resettlement facts. Published October 2023. Accessed April 8, 2024. https://www.unhcr.org/us/media/refugee-resettlement-facts<br/><br/> 4. US Department of State. Report to Congress on Proposed Refugee Admissions for Fiscal Year 2024. Published November 3, 2023. Accessed April 8, 2024. https://www.state.gov/report-to-congress-on-proposed-refugee-admissions-for-fiscal-year-2024/ <br/><br/> 5. UNHCR. Compact for Migration: Definitions. United Nations. Accessed April 4, 2024. https://refugeesmigrants.un.org/definitions<br/><br/> 6. United Nations High Commissioner for Refugees (UNHCR). Convention and Protocol Relating to the Status of Refugees. Published December 2010. Accessed January 11, 2024. https://www.unhcr.org/us/media/convention-and-protocol-relating-status-refugees<br/><br/> 7. Kibar Öztürk M. Skin diseases in rural Nyala, Sudan (in a rural hospital, in 12 orphanages, and in two refugee camps). <i>Int J Dermatol</i>. 2019;58:1341-1349. doi:10.1111/ijd.14619<br/><br/> 8. Padovese V, Knapp A. Challenges of managing skin diseases in refugees and migrants. <i>Dermatol Clin</i>. 2021;39:101-115. doi:10.1016/j.det.2020.08.010<br/><br/> 9. Saikal SL, Ge L, Mir A, et al. Skin disease profile of Syrian refugees in Jordan: a field-mission assessment. <i>J Eur Acad Dermatol Venereol.</i> 2020;34:419-425. doi:10.1111/jdv.15909<br/><br/>10. Eonomopoulou A, Pavli A, Stasinopoulou P, et al. Migrant screening: lessons learned from the migrant holding level at the Greek-Turkish borders. <i>J Infect Public Health</i>. 2017;10:177-184. doi:10.1016/j.jiph.2016.04.012<br/><br/>11. Marano N, Angelo KM, Merrill RD, et al. Expanding travel medicine in the 21st century to address the health needs of the world’s migrants.<i>J Travel Med</i>. 2018;25. doi:10.1093/jtm/tay067<br/><br/>12. Hay RJ, Asiedu K. Skin-related neglected tropical diseases (skin NTDs)—a new challenge. <i>Trop Med Infect Dis</i>. 2018;4. doi:10.3390/tropicalmed4010004<br/><br/>13. NIAID. Neglected tropical diseases. Updated July 11, 2016. Accessed April 4, 2024. https://www.niaid.nih.gov/research/neglected-tropical-diseases<br/><br/>14. Arlian LG, Morgan MS. A review of Sarcoptes scabiei: past, present and future. <i>Parasit Vectors</i>. 2017;10:297. doi:10.1186/s13071-017-2234-1<br/><br/>15. Arlian LG, Runyan RA, Achar S, et al. Survival and infectivity of Sarcoptes scabiei var. canis and var. hominis. <i>J Am Acad Dermatol</i>. 1984;11(2 pt 1):210-215. doi:10.1016/s0190-9622(84)70151-4<br/><br/>16. Chandler DJ, Fuller LC. A review of scabies: an infestation more than skin deep. <i>Dermatology</i>. 2019;235:79-90. doi:10.1159/000495290<br/><br/>17. Karimkhani C, Colombara DV, Drucker AM, et al. The global burden of scabies: a cross-sectional analysis from the Global Burden of Disease Study 2015. <i>Lancet Infect Dis</i>. 2017;17:1247-1254. doi:10.1016/S1473-3099(17)30483-8<br/><br/>18. Romani L, Steer AC, Whitfeld MJ, et al. Prevalence of scabies and impetigo worldwide: a systematic review. <i>Lancet Infect Dis</i>. 2015;15:960-967. doi:10.1016/S1473-3099(15)00132-2<br/><br/>19. Thomas C, Coates SJ, Engelman D, et al. Ectoparasites: scabies. <i>J Am Acad Dermatol</i>. 2020;82:533-548. doi:10.1016/j.jaad.2019.05.109<br/><br/>20. Mellanby K, Johnson CG, Bartley WC. Treatment of scabies. <i>Br Med J</i>. 1942;2:1-4. doi:10.1136/bmj.2.4252.1<br/><br/>21. Walton SF. The immunology of susceptibility and resistance to scabies. <i>Parasit Immunol</i>. 2010;32:532-540. doi:10.1111/j.1365-3024.2010.01218.x<br/><br/>22. Coates SJ, Thomas C, Chosidow O, et al. Ectoparasites: pediculosis and tungiasis. <i>J Am Acad Dermatol</i>. 2020;82:551-569. doi:10.1016/j.jaad.2019.05.110</p> <p class="reference">23. Engelman D, Fuller LC, Steer AC; International Alliance for the Control of Scabies Delphi p. Consensus criteria for the diagnosis of scabies: a Delphi study of international experts. <i>PLoS Negl Trop Dis</i>. 2018;12:E0006549. doi:10.1371/journal.pntd.0006549<br/><br/>24. World Health Organization. WHO Model Lists of Essential Medicines—23rd list, 2023. Updated July 26, 2023. Accessed April 8, 2024. https://www.who.int/publications/i/item/WHO-MHP-HPS-EML-2023.02 <br/><br/>25. Salavastru CM, Chosidow O, Boffa MJ, et al. European guideline for the management of scabies. <i>J Eur Acad Dermatol Venereol</i>. 2017;31:1248-1253. doi:10.1111/jdv.14351<br/><br/>26. Badiaga S, Brouqui P. Human louse-transmitted infectious diseases. <i>Clin Microbiol Infect</i>. 2012;18:332-337. doi:10.1111/j.1469-0691.2012.03778.x<br/><br/>27. Leo NP, Campbell NJH, Yang X, et al. Evidence from mitochondrial DNA that head lice and body lice of humans (Phthiraptera: Pediculidae) are conspecific. <i>J Med Entomol. </i>2002;39:662-666. doi:10.1603/0022-2585-39.4.662<br/><br/>28. Chosidow O. Scabies and pediculosis. <i>Lancet</i>. 2000;355:819-826. doi:10.1016/S0140-6736(99)09458-1<br/><br/>29. Arnaud A, Chosidow O, Détrez M-A, et al. Prevalences of scabies and pediculosis corporis among homeless people in the Paris region: results from two randomized cross-sectional surveys (HYTPEAC study). <i>Br J Dermatol</i>. 2016;174:104-112. doi:10.1111/bjd.14226<br/><br/>30. Brouqui P. Arthropod-borne diseases associated with political and social disorder. <i>Annu Rev Entomol</i>. 2011;56:357-374. doi:10.1146/annurev-ento-120709-144739<br/><br/>31. Ko CJ, Elston DM. Pediculosis. <i>J Am Acad Dermatol</i>. 2004;50:1-12. doi:10.1016/S0190-9622(03)02729-4<br/><br/>32. Bloomfield D. Head lice. <i>Pediatr Rev</i>. 2002;23:34-35; discussion 34-35. doi:10.1542/pir.23-1-34<br/><br/>33. Stone SP GJ, Bacelieri RE. Scabies, other mites, and pediculosis. In: Wolf K GL, Katz SI, et al (eds). <i>Fitzpatrick’s Dermatology in General Medicine</i>. McGraw Hill; 2008:2029.<br/><br/>34. Foucault C, Ranque S, Badiaga S, et al. Oral ivermectin in the treatment of body lice. <i>J Infect Dis</i>. 2006;193:474-476. doi:10.1086/499279<br/><br/>35. Benkouiten S, Drali R, Badiaga S, et al. Effect of permethrin-impregnated underwear on body lice in sheltered homeless persons: a randomized controlled trial. <i>JAMA Dermatol. </i>2014;150:273-279. doi:10.1001/jamadermatol.2013.6398<br/><br/>36. CDC. Parasites: Treatment. Updated October 15, 2019. Accessed April 4, 2024. https://www.cdc.gov/parasites/lice/head/treatment.html<br/><br/>37. Devore CD, Schutze GE; Council on School Health and Committee on Infectious Diseases, American Academy of Pediatrics. Head lice. <i>Pediatrics</i>. 2015;135:e1355-e1365. doi:10.1542/peds.2015-0746<br/><br/>38. Ohl ME, Spach DH. Bartonella quintana and urban trench fever. <i>Clin Infect Dis</i>. 2000;31:131-135. doi:10.1086/313890<br/><br/>39. Drali R, Sangaré AK, Boutellis A, et al. Bartonella quintana in body lice from scalp hair of homeless persons, France. <i>Emerg Infect Dis</i>. 2014;20:907-908. doi:10.3201/eid2005.131242<br/><br/>40. Rudd N, Zakaria A, Kohn MA, et al. Association of body lice infestation with hemoglobin values in hospitalized dermatology patients. <i>JAMA Dermatol</i>. 2022;158:691-693. doi:10.1001/jamadermatol.2022.0818</p> <p class="reference">41. Guss DA, Koenig M, Castillo EM. Severe iron deficiency anemia and lice infestation. <i>J Emergency Med</i>. 2011;41:362-365. doi:10.1016/j.jemermed.2010.05.030<br/><br/>42. Neglected tropical diseases of the skin: WHO launches mobile application to facilitate diagnosis. News release. World Health Organization; July 16, 2020. Accessed April 4, 2024. https://www.who.int/news/item/16-07-2020-neglected-tropical-diseases-of-the-skin-who-launches-mobile-application-to-facilitate-diagnosis<br/><br/>43. Padovese V, Fuller LC, Griffiths CEM, et al; Migrant Health Dermatology Working Group of the International Foundation for Dermatology. Migrant skin health: perspectives from the Migrant Health Summit, Malta, 2022. <i>Br J Dermatology</i>. 2023;188:553-554. doi:10.1093/bjd/ljad001 <br/><br/>44. Knapp AP, Rehmus W, Chang AY. Skin diseases in displaced populations: a review of contributing factors, challenges, and approaches to care. <i>Int J Dermato</i>l. 2020;59:1299-1311. doi:10.1111/ijd.15063<br/><br/>45. Norman FF, Comeche B, Chamorro S, et al. Overcoming challenges in the diagnosis and treatment of parasitic infectious diseases in migrants. <i>Expert Rev Anti-infective Therapy. </i>2020;18:127-143. doi:10.1080/14787210.2020.1713099<br/><br/>46. Skin NTDs: prioritizing integrated approaches to reduce suffering, psychosocial impact and stigmatization. News release. World Health Organization; October 29, 2020. Accessed April 4, 2024. https://www.who.int/news/item/29-10-2020-skin-ntds-prioritizing-integrated-approaches-to-reduce-suffering-psychosocial-impact-and-stigmatization</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Alexis G. Strahan is from the Mercer University School of Medicine, Savannah, Georgia. Dr. Elston is from the Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>All images are in the public domain. <br/><br/>Correspondence: Alexis G. Strahan, MD, MSN, 55 Fruit St, Bartlett Hall 6R, Boston, MA 02114 (<a href="mailto:alexis.grabow.strahan@live.mercer.edu">alexis.grabow.strahan@live.mercer.edu</a>). <br/><br/><em><hl name="17868"/>Cutis. </em>2024 April;113(4):E16-E21. doi:10.12788/cutis.0999 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>War and natural disasters displace populations and disrupt infrastructure and access to medical care.</li> <li>Infestations and cutaneous infections are common among refugee populations, and impetigo often is a sign of underlying scabies infestation.</li> <li>Body lice are important disease vectors inrefugee populations. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • War and natural disasters displace populations and disrupt infrastructure and access to medical care.
  • Infestations and cutaneous infections are common among refugee populations, and impetigo often is a sign of underlying scabies infestation.
  • Body lice are important disease vectors inrefugee populations.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Evaluating the Cost Burden of Alopecia Areata Treatment: A Comprehensive Review for Dermatologists

Article Type
Changed
Wed, 04/10/2024 - 10:10
Display Headline
Evaluating the Cost Burden of Alopecia Areata Treatment: A Comprehensive Review for Dermatologists

Alopecia areata (AA) affects 4.5 million individuals in the United States, with 66% younger than 30 years.1,2 Inflammation causes hair loss in well-circumscribed, nonscarring patches on the body with a predilection for the scalp.3-6 The disease can devastate a patient’s self-esteem, in turn reducing quality of life.1,7 Alopecia areata is an autoimmune T-cell–mediated disease in which hair follicles lose their immune privilege.8-10 Several specific mechanisms in the cytokine interactions between T cells and the hair follicle have been discovered, revealing the Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathway as pivotal in the pathogenesis of the disease and leading to the use of JAK inhibitors for treatment.11

There is no cure for AA, and the condition is managed with prolonged medical treatments and cosmetic therapies.2 Although some patients may be able to manage the annual cost, the cumulative cost of AA treatment can be burdensome.12 This cumulative cost may increase if newer, potentially expensive treatments become the standard of care. Patients with AA report dipping into their savings (41.3%) and cutting back on food or clothing expenses (33.9%) to account for the cost of alopecia treatment. Although prior estimates of the annual out-of-pocket cost of AA treatments range from $1354 to $2685, the cost burden of individual therapies is poorly understood.12-14

Patients who must juggle expensive medical bills with basic living expenses may be lost to follow-up or fall into treatment nonadherence.15 Other patients’ out-of-pocket costs may be manageable, but the costs to the health care system may compromise care in other ways. We conducted a literature review of the recommended therapies for AA based on American Academy of Dermatology (AAD) guidelines to identify the costs of alopecia treatment and consolidate the available data for the practicing dermatologist.

Methods

We conducted a PubMed search of articles indexed for MEDLINE through September 15, 2022, using the terms alopecia and cost plus one of the treatments (n=21) identified by the AAD2 for the treatment of AA (Figure). The reference lists of included articles were reviewed to identify other potentially relevant studies. Forty-five articles were identified.

Patel_Figure.jpg
%3Cp%3ELiterature%20review%20methodology%20on%20costs%20of%20alopecia%20areata%20(AA)%20treatment.%20JAK%20indicates%20Janus%20kinase.%3C%2Fp%3E

Given the dearth of cost research in alopecia and the paucity of large prospective studies, we excluded articles that were not available in their full-text form or were not in English (n=3), articles whose primary study topic was not AA or an expert-approved alopecia treatment (n=15), and articles with no concrete cost data (n=17), which yielded 10 relevant articles that we studied using qualitative analysis.

Due to substantial differences in study methods and outcome measures, we did not compare the costs of alopecia among studies and did not perform statistical analysis. The quality of each study was investigated and assigned a level of evidence per the 2009 criteria from the Centre for Evidence-Based Medicine.16

 

 

All cost data were converted into US dollars ($) using the conversion rate from the time of the original article’s publication.

Results

Total and Out-of-pocket Costs of AA—Li et al13 studied out-of-pocket health care costs for AA patients (N=675). Of these participants, 56.9% said their AA was moderately to seriously financially burdensome, and 41.3% reported using their savings to manage these expenses. Participants reported median out-of-pocket spending of $1354 (interquartile range, $537–$3300) annually. The most common categories of expenses were hair appointments (81.8%) and vitamins/supplements (67.7%).13

Mesinkovska et al14 studied the qualitative and quantitative financial burdens of moderate to severe AA (N=216). Fifty-seven percent of patients reported the financial impact of AA as moderately to severely burdensome with a willingness to borrow money or use savings to cover out-of-pocket costs. Patients without insurance cited cost as a major barrier to obtaining reatment. In addition to direct treatment-related expenses, AA patients spent a mean of $1961 per year on therapy to cope with the disease’s psychological burden. Lost work hours represented another source of financial burden; 61% of patients were employed, and 45% of them reported missing time from their job because of AA.14

Mostaghimi et al12 studied health care resource utilization and all-cause direct health care costs in privately insured AA patients with or without alopecia totalis (AT) or alopecia universalis (AU)(n=14,972) matched with non-AA controls (n=44,916)(1:3 ratio). Mean total all-cause medical and pharmacy costs were higher in both AA groups compared with controls (AT/AU, $18,988 vs $11,030; non-AT/AU, $13,686 vs $9336; P<.001 for both). Out-of-pocket costs were higher for AA vs controls (AT/AU, $2685 vs $1457; non-AT/AU, $2223 vs $1341; P<.001 for both). Medical costs in the AT/AU and non-AT/AU groups largely were driven by outpatient costs (AT/AU, $10,277 vs $5713; non-AT/AU, $8078 vs $4672; P<.001 for both).12

Costs of Concealment—When studying the out-of-pocket costs of AA (N=675), Li et al13 discovered that the median yearly spending was highest on headwear or cosmetic items such as hats, wigs, and makeup ($450; interquartile range, $50–$1500). Mesinkovska et al14 reported that 49% of patients had insurance that covered AA treatment. However, 75% of patients reported that their insurance would not cover costs of concealment (eg, weave, wig, hair piece). Patients (N=112) spent a mean of $2211 per year and 10.3 hours per week on concealment.14

Minoxidil—Minoxidil solution is available over-the-counter, and its ease of access makes it a popular treatment for AA.17 Because manufacturers can sell directly to the public, minoxidil is marketed with bold claims and convincing packaging. Shrank18 noted that the product can take 4 months to work, meaning customers must incur a substantial cost burden before realizing the treatment’s benefit, which is not always obvious when purchasing minoxidil products, leaving customers—who were marketed a miracle drug—disappointed. Per Shrank,18 patients who did not experience hair regrowth after 4 months were advised to continue treatment for a year, leading them to spend hundreds of dollars for uncertain results. Those who did experience hair regrowth were advised to continue using the product twice daily 7 days per week indefinitely.18

Wehner et al19 studied the association between gender and drug cost for over-the-counter minoxidil. The price that women paid for 2% regular-strength minoxidil solutions was similar to the price that men paid for 5% extra-strength minoxidil solutions (women’s 2%, $7.63/30 mL; men’s 5%, $7.61/30 mL; P=.67). Minoxidil 5% foams with identical ingredients were priced significantly more per volume of the same product when sold as a product directed at women vs a product directed at men (men’s 5%, $8.05/30 mL; women’s 5%, $11.27/30 mL; P<.001).19

 

 

Beach20 compared the cost of oral minoxidil to topical minoxidil. At $28.60 for a 3-month supply, oral minoxidil demonstrated cost savings compared to topical minoxidil ($48.30).20

Diphencyprone—Bhat et al21 studied the cost-efficiency of diphencyprone (DPC) in patients with AA resistant to at least 2 conventional treatments (N=29). After initial sensitization with 2% DPC, patients received weekly or fortnightly treatments. Most of the annual cost burden of DPC treatment was due to staff time and overhead rather than the cost of the DPC itself: $258 for the DPC, $978 in staff time and overhead for the department, and $1233 directly charged to the patient.21

Lekhavat et al22 studied the economic impact of home-use vs office-use DPC in extensive AA (N=82). Both groups received weekly treatments in the hospital until DPC concentrations had been adjusted. Afterward, the home group was given training on self-applying DPC at home. The home group had monthly office visits for DPC concentration evaluation and refills, while the office group had weekly appointments for DPC treatment at the hospital. Calculated costs included those to the health care provider (ie, material, labor, capital costs) and the patient’s final out-of-pocket expense. The total cost to the health care provider was higher for the office group than the home group at 48 weeks (office, $683.52; home, $303.67; P<.001). Median out-of-pocket costs did not vary significantly between groups, which may have been due to small sample size affecting the range (office, $418.07; home, $189.69; P=.101). There was no significant difference between groups in the proportion of patients who responded favorably to the DPC.22

JAK Inhibitors—Chen et al23 studied the efficacy of low-dose (5 mg) tofacitinib to treat severe AA (N=6). Compared to prior studies,24-27 this analysis reported the efficacy of low-dose tofacitinib was not inferior to higher doses (10–20 mg), and low-dose tofacitinib reduced treatment costs by more than 50%.23

Per the GlobalData Healthcare database, the estimated annual cost of therapy for JAK inhibitors following US Food and Drug Administration approval was $50,000. At the time of their reporting, the next most expensive immunomodulatory drug for AA was cyclosporine, with an annual cost of therapy of $1400.28 Dillon29 reviewed the use of JAK inhibitors for the treatment of AA. The cost estimates by Dillon29 prior to FDA approval aligned with the pricing of Eli Lilly and Company for the now-approved JAK inhibitor baricitinib.30 The list price of baricitinib is $2739.99 for a 30-day supply of 2-mg tablets or $5479.98 for a 30-day supply of 4-mg tablets. This amounts to $32,879.88 for an annual supply of 2-mg tablets and $65,759.76 for an annual supply for 4-mg tablets, though the out-of-pocket costs will vary.30

Comment

We reviewed the global and treatment-specific costs of AA, consolidating the available data for the practicing dermatologist. Ten studies of approximately 16,000 patients with AA across a range of levels of evidence (1a to 4) were included (Table). Three of 10 articles studied global costs of AA, 1 studied costs of concealment, 3 studied costs of minoxidil, 2 studied costs of DPC, and 2 studied costs of JAK inhibitors. Only 2 studies achieved level of evidence 1a: the first assessed the economic impact of home-use vs office-use DPC,22 and the second researched the efficacy and outcomes of JAK inhibitors.29

CT113004185_Table_part1.jpg

CT113004185_Table_part2.jpg

Hair-loss treatments and concealment techniques cost the average patient thousands of dollars. Spending was highest on headwear or cosmetic items, which were rarely covered by insurance.13 Psychosocial sequelae further increased cost via therapy charges and lost time at work.14 Patients with AA had greater all-cause medical costs than those without AA, with most of the cost driven by outpatient visits. Patients with AA also paid nearly twice as much as non-AA patients on out-of-pocket health care expenses.14 Despite the high costs and limited efficacy of many AA therapies, patients reported willingness to incur debt or use savings to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress.13

 

 

Minoxidil solution does not require physician office visits and is available over-the-counter.17 Despite identical ingredients, minoxidil is priced more per volume when marketed to women compared with men, which reflects the larger issue of gender-based pricing that does not exist for other AAD-approved alopecia therapies but may exist for cosmetic treatments and nonapproved therapies (eg, vitamins/supplements) that are popular in the treatment of AA.19 Oral minoxidil was more cost-effective than the topical form, and gender-based pricing was a nonissue.20 However, oral minoxidil requires a prescription, mandating patients incur the cost of an office visit. Patients should be wary of gender- or marketing-related surcharges for minoxidil solutions, and oral minoxidil may be a cost-effective choice.

Diphencyprone is a relatively affordable drug for AA, but the regular office visits traditionally required for its administration increase associated cost.21 Self-administration of DPC at home was more cost- and time-effective than in-office DPC administration and did not decrease efficacy. A regimen combining office visits for initial DPC titration, at-home DPC administration, and periodic office follow-up could minimize costs while preserving outcomes and safety.22

Janus kinase inhibitors are cutting-edge and expensive therapies for AA. The annual cost of these medications poses a tremendous burden on the payer (list price of annual supply ritlecitinib is $49,000),31 be that the patient or the insurance company. Low-dose tofacitinib may be similarly efficacious and could substantially reduce treatment costs.23 The true utility of these medications, specifically considering their steep costs, remains to be determined.

Conclusion

Alopecia areata poses a substantial and recurring cost burden on patients that is multifactorial including treatment, office visits, concealment, alternative therapies, psychosocial costs, and missed time at work. Although several treatment options exist, none of them are definitive. Oral minoxidil and at-home DPC administration can be cost-effective, though the cumulative cost is still high. The cost utility of JAK inhibitors remains unclear. When JAK inhibitors are prescribed, low-dose therapy may be used as maintenance to curb treatment costs. Concealment and therapy costs pose an additional, largely out-of-pocket financial burden. Despite the limited efficacy of many AA therapies, patients incur substantial expenses to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress. There are no head-to-head studies comparing the cost-effectiveness of the different AA therapies; thus, it is unclear if one treatment is most efficacious. This topic remains an avenue for future investigation. Much of the cost burden of AA treatment falls directly on patients. Increasing coverage of AA-associated expenses, such as minoxidil therapy or wigs, could decrease the cost burden on patients. Providers also can inform patients about cost-saving tactics, such as purchasing minoxidil based on concentration and vehicle rather than marketing directed at men vs women. Finally, some patients may have insurance plans that at least partially cover the costs of wigs but may not be aware of this benefit. Querying a patient’s insurance provider can further minimize costs.

References
  1. Tosti A, Piraccini BM, Pazzaglia M, et al. Clobetasol propionate 0.05% under occlusion in the treatment of alopecia totalis/universalis. J Am Acad Dermatol. 2003;49:96-98. doi:10.1067/mjd.2003.423
  2. Strazzulla LC, Wang EHC, Avila L, et al. Alopecia areata: an appraisal of new treatment approaches and overview of current therapies. J Am Acad Dermatol. 2018;78:15-24. doi:10.1016/j.jaad.2017.04.1142
  3. Olsen EA, Carson SC, Turney EA. Systemic steroids with or without 2% topical minoxidil in the treatment of alopecia areata. Arch Dermatol. 1992;128:1467-1473.
  4. Levy LL, Urban J, King BA. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. J Am Acad Dermatol. 2015;73:395-399. doi:10.1016/j.jaad.2015.06.045
  5. Ports WC, Khan S, Lan S, et al. A randomized phase 2a efficacy and safety trial of the topical Janus kinase inhibitor tofacitinib in the treatment of chronic plaque psoriasis. Br J Dermatol. 2013;169:137-145. doi:10.1111/bjd.12266
  6. Strober B, Buonanno M, Clark JD, et al. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol. 2013;169:992-999. doi:10.1111/bjd.12517
  7. van der Steen PH, van Baar HM, Happle R, et al. Prognostic factors in the treatment of alopecia areata with diphenylcyclopropenone. J Am Acad Dermatol. 1991;24(2, pt 1):227-230. doi:10.1016/0190-9622(91)70032-w
  8. Strazzulla LC, Avila L, Lo Sicco K, et al. Image gallery: treatment of refractory alopecia universalis with oral tofacitinib citrate and adjunct intralesional triamcinolone injections. Br J Dermatol. 2017;176:E125. doi:10.1111/bjd.15483
  9. Madani S, Shapiro J. Alopecia areata update. J Am Acad Dermatol. 2000;42:549-566; quiz 567-570.
  10. Carnahan MC, Goldstein DA. Ocular complications of topical, peri-ocular, and systemic corticosteroids. Curr Opin Ophthalmol. 2000;11:478-483. doi:10.1097/00055735-200012000-00016
  11. Harel S, Higgins CA, Cerise JE, et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. Sci Adv. 2015;1:E1500973. doi:10.1126/sciadv.1500973
  12. Mostaghimi A, Gandhi K, Done N, et al. All-cause health care resource utilization and costs among adults with alopecia areata: a retrospective claims database study in the United States. J Manag Care Spec Pharm. 2022;28:426-434. doi:10.18553/jmcp.2022.28.4.426
  13. Li SJ, Mostaghimi A, Tkachenko E, et al. Association of out-of-pocket health care costs and financial burden for patients with alopecia areata. JAMA Dermatol. 2019;155:493-494. doi:10.1001/jamadermatol.2018.5218
  14. Mesinkovska N, King B, Mirmirani P, et al. Burden of illness in alopecia areata: a cross-sectional online survey study. J Investig Dermatol Symp Proc. 2020;20:S62-S68. doi:10.1016/j.jisp.2020.05.007
  15. Iuga AO, McGuire MJ. Adherence and health care costs. Risk Manag Healthc Policy. 2014;7:35-44. doi:10.2147/rmhp.S19801
  16. Oxford Centre for Evidence-Based Medicine: Levels of Evidence (March 2009). University of Oxford website. Accessed March 25, 2024. https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009
  17. Klifto KM, Othman S, Kovach SJ. Minoxidil, platelet-rich plasma (PRP), or combined minoxidil and PRP for androgenetic alopecia in men: a cost-effectiveness Markov decision analysis of prospective studies. Cureus. 2021;13:E20839. doi:10.7759/cureus.20839
  18. Shrank AB. Minoxidil over the counter. BMJ. 1995;311:526. doi:10.1136/bmj.311.7004.526
  19. Wehner MR, Nead KT, Lipoff JB. Association between gender and drug cost for over-the-counter minoxidil. JAMA Dermatol. 2017;153:825-826.
  20. Beach RA. Case series of oral minoxidil for androgenetic and traction alopecia: tolerability & the five C’s of oral therapy. Dermatol Ther. 2018;31:E12707. doi:10.1111/dth.12707
  21. Bhat A, Sripathy K, Wahie S, et al. Efficacy and cost-efficiency of diphencyprone for alopecia areata. Br J Dermatol. 2011;165:43-44.
  22. Lekhavat C, Rattanaumpawan P, Juengsamranphong I. Economic impact of home-use versus office-use diphenylcyclopropenone in extensive alopecia areata. Skin Appendage Disord. 2022;8:108-117.
  23. Chen YY, Lin SY, Chen YC, et al. Low-dose tofacitinib for treating patients with severe alopecia areata: an efficient and cost-saving regimen. Eur J Dermatol. 2019;29:667-669. doi:10.1684/ejd.2019.3668
  24. Liu LY, Craiglow BG, Dai F, et al. Tofacitinib for the treatment of severe alopecia areata and variants: a study of 90 patients. J Am Acad Dermatol. 2017;76:22-28. doi:10.1016/j.jaad.2016.09.007
  25. Kennedy Crispin M, Ko JM, Craiglow BG, et al. Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with alopecia areata. JCI Insight. 2016;1:e89776. doi:10.1172/jci.insight.89776
  26. Jabbari A, Sansaricq F, Cerise J, et al. An open-label pilot study to evaluate the efficacy of tofacitinib in moderate to severe patch-type alopecia areata, totalis, and universalis. J Invest Dermatol. 2018;138:1539-1545. doi:10.1016/j.jid.2018.01.032
  27. Craiglow BG, Liu LY, King BA. Tofacitinib for the treatment of alopecia areata and variants in adolescents. J Am Acad Dermatol. 2017;76:29-32. doi:10.1016/j.jaad.2016.09.006
  28. GlobalData Healthcare. Can JAK inhibitors penetrate the alopecia areata market effectively? Pharmaceutical Technology. July 15, 2019. Accessed February 8, 2024. https://www.pharmaceutical-technology.com/analyst-comment/alopecia-areata-treatment-2019/
  29. Dillon KL. A comprehensive literature review of JAK inhibitors in treatment of alopecia areata. Clin Cosmet Investig Dermatol. 2021;14:691-714. doi:10.2147/ccid.S309215
  30. How much should I expect to pay for Olumiant? Accessed March 20, 2024. https://www.lillypricinginfo.com/olumiant
  31. McNamee A. FDA approves first-ever adolescent alopecia treatment from Pfizer. Pharmaceutical Technology. June 26, 2023. Accessed March 20, 2024. https://www.pharmaceutical-technology.com/news/fda-approves-first-ever-adolescent-alopecia-treatment-from-pfizer/?cf-view
Article PDF
Author and Disclosure Information

Palak V. Patel, Angelica Coello, and Dr. McMichael are from the Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Larrondo is from the Department of Dermatology, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.

Palak V. Patel, Angelica Coello, and Dr. Larrondo report no conflict of interest. Dr. McMichael has received research, speaking, and/or consulting support from AbbVie; Arcutis Biotherapeutics; Bristol Meyers Squibb; Concert Pharmaceuticals, Inc; Eli Lilly and Company; eResearch Technology, Inc; Galderma; Incyte Corporation; Informa Healthcare; Janssen Pharmaceuticals; Johnson & Johnson; L’Oréal; Pfizer; Procter and Gamble; REVIAN, Inc; Samumed; Sanofi-Regeneron; Sun Pharmaceuticls; and UCB.

Correspondence: Palak V. Patel, BA, BS, 1 Medical Center Blvd, Winston-Salem, NC 27157-1071 (palpatel@wakehealth.edu).

Publications
Topics
Page Number
185-190
Sections
Author and Disclosure Information

Palak V. Patel, Angelica Coello, and Dr. McMichael are from the Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Larrondo is from the Department of Dermatology, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.

Palak V. Patel, Angelica Coello, and Dr. Larrondo report no conflict of interest. Dr. McMichael has received research, speaking, and/or consulting support from AbbVie; Arcutis Biotherapeutics; Bristol Meyers Squibb; Concert Pharmaceuticals, Inc; Eli Lilly and Company; eResearch Technology, Inc; Galderma; Incyte Corporation; Informa Healthcare; Janssen Pharmaceuticals; Johnson & Johnson; L’Oréal; Pfizer; Procter and Gamble; REVIAN, Inc; Samumed; Sanofi-Regeneron; Sun Pharmaceuticls; and UCB.

Correspondence: Palak V. Patel, BA, BS, 1 Medical Center Blvd, Winston-Salem, NC 27157-1071 (palpatel@wakehealth.edu).

Author and Disclosure Information

Palak V. Patel, Angelica Coello, and Dr. McMichael are from the Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Larrondo is from the Department of Dermatology, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.

Palak V. Patel, Angelica Coello, and Dr. Larrondo report no conflict of interest. Dr. McMichael has received research, speaking, and/or consulting support from AbbVie; Arcutis Biotherapeutics; Bristol Meyers Squibb; Concert Pharmaceuticals, Inc; Eli Lilly and Company; eResearch Technology, Inc; Galderma; Incyte Corporation; Informa Healthcare; Janssen Pharmaceuticals; Johnson & Johnson; L’Oréal; Pfizer; Procter and Gamble; REVIAN, Inc; Samumed; Sanofi-Regeneron; Sun Pharmaceuticls; and UCB.

Correspondence: Palak V. Patel, BA, BS, 1 Medical Center Blvd, Winston-Salem, NC 27157-1071 (palpatel@wakehealth.edu).

Article PDF
Article PDF

Alopecia areata (AA) affects 4.5 million individuals in the United States, with 66% younger than 30 years.1,2 Inflammation causes hair loss in well-circumscribed, nonscarring patches on the body with a predilection for the scalp.3-6 The disease can devastate a patient’s self-esteem, in turn reducing quality of life.1,7 Alopecia areata is an autoimmune T-cell–mediated disease in which hair follicles lose their immune privilege.8-10 Several specific mechanisms in the cytokine interactions between T cells and the hair follicle have been discovered, revealing the Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathway as pivotal in the pathogenesis of the disease and leading to the use of JAK inhibitors for treatment.11

There is no cure for AA, and the condition is managed with prolonged medical treatments and cosmetic therapies.2 Although some patients may be able to manage the annual cost, the cumulative cost of AA treatment can be burdensome.12 This cumulative cost may increase if newer, potentially expensive treatments become the standard of care. Patients with AA report dipping into their savings (41.3%) and cutting back on food or clothing expenses (33.9%) to account for the cost of alopecia treatment. Although prior estimates of the annual out-of-pocket cost of AA treatments range from $1354 to $2685, the cost burden of individual therapies is poorly understood.12-14

Patients who must juggle expensive medical bills with basic living expenses may be lost to follow-up or fall into treatment nonadherence.15 Other patients’ out-of-pocket costs may be manageable, but the costs to the health care system may compromise care in other ways. We conducted a literature review of the recommended therapies for AA based on American Academy of Dermatology (AAD) guidelines to identify the costs of alopecia treatment and consolidate the available data for the practicing dermatologist.

Methods

We conducted a PubMed search of articles indexed for MEDLINE through September 15, 2022, using the terms alopecia and cost plus one of the treatments (n=21) identified by the AAD2 for the treatment of AA (Figure). The reference lists of included articles were reviewed to identify other potentially relevant studies. Forty-five articles were identified.

Patel_Figure.jpg
%3Cp%3ELiterature%20review%20methodology%20on%20costs%20of%20alopecia%20areata%20(AA)%20treatment.%20JAK%20indicates%20Janus%20kinase.%3C%2Fp%3E

Given the dearth of cost research in alopecia and the paucity of large prospective studies, we excluded articles that were not available in their full-text form or were not in English (n=3), articles whose primary study topic was not AA or an expert-approved alopecia treatment (n=15), and articles with no concrete cost data (n=17), which yielded 10 relevant articles that we studied using qualitative analysis.

Due to substantial differences in study methods and outcome measures, we did not compare the costs of alopecia among studies and did not perform statistical analysis. The quality of each study was investigated and assigned a level of evidence per the 2009 criteria from the Centre for Evidence-Based Medicine.16

 

 

All cost data were converted into US dollars ($) using the conversion rate from the time of the original article’s publication.

Results

Total and Out-of-pocket Costs of AA—Li et al13 studied out-of-pocket health care costs for AA patients (N=675). Of these participants, 56.9% said their AA was moderately to seriously financially burdensome, and 41.3% reported using their savings to manage these expenses. Participants reported median out-of-pocket spending of $1354 (interquartile range, $537–$3300) annually. The most common categories of expenses were hair appointments (81.8%) and vitamins/supplements (67.7%).13

Mesinkovska et al14 studied the qualitative and quantitative financial burdens of moderate to severe AA (N=216). Fifty-seven percent of patients reported the financial impact of AA as moderately to severely burdensome with a willingness to borrow money or use savings to cover out-of-pocket costs. Patients without insurance cited cost as a major barrier to obtaining reatment. In addition to direct treatment-related expenses, AA patients spent a mean of $1961 per year on therapy to cope with the disease’s psychological burden. Lost work hours represented another source of financial burden; 61% of patients were employed, and 45% of them reported missing time from their job because of AA.14

Mostaghimi et al12 studied health care resource utilization and all-cause direct health care costs in privately insured AA patients with or without alopecia totalis (AT) or alopecia universalis (AU)(n=14,972) matched with non-AA controls (n=44,916)(1:3 ratio). Mean total all-cause medical and pharmacy costs were higher in both AA groups compared with controls (AT/AU, $18,988 vs $11,030; non-AT/AU, $13,686 vs $9336; P<.001 for both). Out-of-pocket costs were higher for AA vs controls (AT/AU, $2685 vs $1457; non-AT/AU, $2223 vs $1341; P<.001 for both). Medical costs in the AT/AU and non-AT/AU groups largely were driven by outpatient costs (AT/AU, $10,277 vs $5713; non-AT/AU, $8078 vs $4672; P<.001 for both).12

Costs of Concealment—When studying the out-of-pocket costs of AA (N=675), Li et al13 discovered that the median yearly spending was highest on headwear or cosmetic items such as hats, wigs, and makeup ($450; interquartile range, $50–$1500). Mesinkovska et al14 reported that 49% of patients had insurance that covered AA treatment. However, 75% of patients reported that their insurance would not cover costs of concealment (eg, weave, wig, hair piece). Patients (N=112) spent a mean of $2211 per year and 10.3 hours per week on concealment.14

Minoxidil—Minoxidil solution is available over-the-counter, and its ease of access makes it a popular treatment for AA.17 Because manufacturers can sell directly to the public, minoxidil is marketed with bold claims and convincing packaging. Shrank18 noted that the product can take 4 months to work, meaning customers must incur a substantial cost burden before realizing the treatment’s benefit, which is not always obvious when purchasing minoxidil products, leaving customers—who were marketed a miracle drug—disappointed. Per Shrank,18 patients who did not experience hair regrowth after 4 months were advised to continue treatment for a year, leading them to spend hundreds of dollars for uncertain results. Those who did experience hair regrowth were advised to continue using the product twice daily 7 days per week indefinitely.18

Wehner et al19 studied the association between gender and drug cost for over-the-counter minoxidil. The price that women paid for 2% regular-strength minoxidil solutions was similar to the price that men paid for 5% extra-strength minoxidil solutions (women’s 2%, $7.63/30 mL; men’s 5%, $7.61/30 mL; P=.67). Minoxidil 5% foams with identical ingredients were priced significantly more per volume of the same product when sold as a product directed at women vs a product directed at men (men’s 5%, $8.05/30 mL; women’s 5%, $11.27/30 mL; P<.001).19

 

 

Beach20 compared the cost of oral minoxidil to topical minoxidil. At $28.60 for a 3-month supply, oral minoxidil demonstrated cost savings compared to topical minoxidil ($48.30).20

Diphencyprone—Bhat et al21 studied the cost-efficiency of diphencyprone (DPC) in patients with AA resistant to at least 2 conventional treatments (N=29). After initial sensitization with 2% DPC, patients received weekly or fortnightly treatments. Most of the annual cost burden of DPC treatment was due to staff time and overhead rather than the cost of the DPC itself: $258 for the DPC, $978 in staff time and overhead for the department, and $1233 directly charged to the patient.21

Lekhavat et al22 studied the economic impact of home-use vs office-use DPC in extensive AA (N=82). Both groups received weekly treatments in the hospital until DPC concentrations had been adjusted. Afterward, the home group was given training on self-applying DPC at home. The home group had monthly office visits for DPC concentration evaluation and refills, while the office group had weekly appointments for DPC treatment at the hospital. Calculated costs included those to the health care provider (ie, material, labor, capital costs) and the patient’s final out-of-pocket expense. The total cost to the health care provider was higher for the office group than the home group at 48 weeks (office, $683.52; home, $303.67; P<.001). Median out-of-pocket costs did not vary significantly between groups, which may have been due to small sample size affecting the range (office, $418.07; home, $189.69; P=.101). There was no significant difference between groups in the proportion of patients who responded favorably to the DPC.22

JAK Inhibitors—Chen et al23 studied the efficacy of low-dose (5 mg) tofacitinib to treat severe AA (N=6). Compared to prior studies,24-27 this analysis reported the efficacy of low-dose tofacitinib was not inferior to higher doses (10–20 mg), and low-dose tofacitinib reduced treatment costs by more than 50%.23

Per the GlobalData Healthcare database, the estimated annual cost of therapy for JAK inhibitors following US Food and Drug Administration approval was $50,000. At the time of their reporting, the next most expensive immunomodulatory drug for AA was cyclosporine, with an annual cost of therapy of $1400.28 Dillon29 reviewed the use of JAK inhibitors for the treatment of AA. The cost estimates by Dillon29 prior to FDA approval aligned with the pricing of Eli Lilly and Company for the now-approved JAK inhibitor baricitinib.30 The list price of baricitinib is $2739.99 for a 30-day supply of 2-mg tablets or $5479.98 for a 30-day supply of 4-mg tablets. This amounts to $32,879.88 for an annual supply of 2-mg tablets and $65,759.76 for an annual supply for 4-mg tablets, though the out-of-pocket costs will vary.30

Comment

We reviewed the global and treatment-specific costs of AA, consolidating the available data for the practicing dermatologist. Ten studies of approximately 16,000 patients with AA across a range of levels of evidence (1a to 4) were included (Table). Three of 10 articles studied global costs of AA, 1 studied costs of concealment, 3 studied costs of minoxidil, 2 studied costs of DPC, and 2 studied costs of JAK inhibitors. Only 2 studies achieved level of evidence 1a: the first assessed the economic impact of home-use vs office-use DPC,22 and the second researched the efficacy and outcomes of JAK inhibitors.29

CT113004185_Table_part1.jpg

CT113004185_Table_part2.jpg

Hair-loss treatments and concealment techniques cost the average patient thousands of dollars. Spending was highest on headwear or cosmetic items, which were rarely covered by insurance.13 Psychosocial sequelae further increased cost via therapy charges and lost time at work.14 Patients with AA had greater all-cause medical costs than those without AA, with most of the cost driven by outpatient visits. Patients with AA also paid nearly twice as much as non-AA patients on out-of-pocket health care expenses.14 Despite the high costs and limited efficacy of many AA therapies, patients reported willingness to incur debt or use savings to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress.13

 

 

Minoxidil solution does not require physician office visits and is available over-the-counter.17 Despite identical ingredients, minoxidil is priced more per volume when marketed to women compared with men, which reflects the larger issue of gender-based pricing that does not exist for other AAD-approved alopecia therapies but may exist for cosmetic treatments and nonapproved therapies (eg, vitamins/supplements) that are popular in the treatment of AA.19 Oral minoxidil was more cost-effective than the topical form, and gender-based pricing was a nonissue.20 However, oral minoxidil requires a prescription, mandating patients incur the cost of an office visit. Patients should be wary of gender- or marketing-related surcharges for minoxidil solutions, and oral minoxidil may be a cost-effective choice.

Diphencyprone is a relatively affordable drug for AA, but the regular office visits traditionally required for its administration increase associated cost.21 Self-administration of DPC at home was more cost- and time-effective than in-office DPC administration and did not decrease efficacy. A regimen combining office visits for initial DPC titration, at-home DPC administration, and periodic office follow-up could minimize costs while preserving outcomes and safety.22

Janus kinase inhibitors are cutting-edge and expensive therapies for AA. The annual cost of these medications poses a tremendous burden on the payer (list price of annual supply ritlecitinib is $49,000),31 be that the patient or the insurance company. Low-dose tofacitinib may be similarly efficacious and could substantially reduce treatment costs.23 The true utility of these medications, specifically considering their steep costs, remains to be determined.

Conclusion

Alopecia areata poses a substantial and recurring cost burden on patients that is multifactorial including treatment, office visits, concealment, alternative therapies, psychosocial costs, and missed time at work. Although several treatment options exist, none of them are definitive. Oral minoxidil and at-home DPC administration can be cost-effective, though the cumulative cost is still high. The cost utility of JAK inhibitors remains unclear. When JAK inhibitors are prescribed, low-dose therapy may be used as maintenance to curb treatment costs. Concealment and therapy costs pose an additional, largely out-of-pocket financial burden. Despite the limited efficacy of many AA therapies, patients incur substantial expenses to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress. There are no head-to-head studies comparing the cost-effectiveness of the different AA therapies; thus, it is unclear if one treatment is most efficacious. This topic remains an avenue for future investigation. Much of the cost burden of AA treatment falls directly on patients. Increasing coverage of AA-associated expenses, such as minoxidil therapy or wigs, could decrease the cost burden on patients. Providers also can inform patients about cost-saving tactics, such as purchasing minoxidil based on concentration and vehicle rather than marketing directed at men vs women. Finally, some patients may have insurance plans that at least partially cover the costs of wigs but may not be aware of this benefit. Querying a patient’s insurance provider can further minimize costs.

Alopecia areata (AA) affects 4.5 million individuals in the United States, with 66% younger than 30 years.1,2 Inflammation causes hair loss in well-circumscribed, nonscarring patches on the body with a predilection for the scalp.3-6 The disease can devastate a patient’s self-esteem, in turn reducing quality of life.1,7 Alopecia areata is an autoimmune T-cell–mediated disease in which hair follicles lose their immune privilege.8-10 Several specific mechanisms in the cytokine interactions between T cells and the hair follicle have been discovered, revealing the Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathway as pivotal in the pathogenesis of the disease and leading to the use of JAK inhibitors for treatment.11

There is no cure for AA, and the condition is managed with prolonged medical treatments and cosmetic therapies.2 Although some patients may be able to manage the annual cost, the cumulative cost of AA treatment can be burdensome.12 This cumulative cost may increase if newer, potentially expensive treatments become the standard of care. Patients with AA report dipping into their savings (41.3%) and cutting back on food or clothing expenses (33.9%) to account for the cost of alopecia treatment. Although prior estimates of the annual out-of-pocket cost of AA treatments range from $1354 to $2685, the cost burden of individual therapies is poorly understood.12-14

Patients who must juggle expensive medical bills with basic living expenses may be lost to follow-up or fall into treatment nonadherence.15 Other patients’ out-of-pocket costs may be manageable, but the costs to the health care system may compromise care in other ways. We conducted a literature review of the recommended therapies for AA based on American Academy of Dermatology (AAD) guidelines to identify the costs of alopecia treatment and consolidate the available data for the practicing dermatologist.

Methods

We conducted a PubMed search of articles indexed for MEDLINE through September 15, 2022, using the terms alopecia and cost plus one of the treatments (n=21) identified by the AAD2 for the treatment of AA (Figure). The reference lists of included articles were reviewed to identify other potentially relevant studies. Forty-five articles were identified.

Patel_Figure.jpg
%3Cp%3ELiterature%20review%20methodology%20on%20costs%20of%20alopecia%20areata%20(AA)%20treatment.%20JAK%20indicates%20Janus%20kinase.%3C%2Fp%3E

Given the dearth of cost research in alopecia and the paucity of large prospective studies, we excluded articles that were not available in their full-text form or were not in English (n=3), articles whose primary study topic was not AA or an expert-approved alopecia treatment (n=15), and articles with no concrete cost data (n=17), which yielded 10 relevant articles that we studied using qualitative analysis.

Due to substantial differences in study methods and outcome measures, we did not compare the costs of alopecia among studies and did not perform statistical analysis. The quality of each study was investigated and assigned a level of evidence per the 2009 criteria from the Centre for Evidence-Based Medicine.16

 

 

All cost data were converted into US dollars ($) using the conversion rate from the time of the original article’s publication.

Results

Total and Out-of-pocket Costs of AA—Li et al13 studied out-of-pocket health care costs for AA patients (N=675). Of these participants, 56.9% said their AA was moderately to seriously financially burdensome, and 41.3% reported using their savings to manage these expenses. Participants reported median out-of-pocket spending of $1354 (interquartile range, $537–$3300) annually. The most common categories of expenses were hair appointments (81.8%) and vitamins/supplements (67.7%).13

Mesinkovska et al14 studied the qualitative and quantitative financial burdens of moderate to severe AA (N=216). Fifty-seven percent of patients reported the financial impact of AA as moderately to severely burdensome with a willingness to borrow money or use savings to cover out-of-pocket costs. Patients without insurance cited cost as a major barrier to obtaining reatment. In addition to direct treatment-related expenses, AA patients spent a mean of $1961 per year on therapy to cope with the disease’s psychological burden. Lost work hours represented another source of financial burden; 61% of patients were employed, and 45% of them reported missing time from their job because of AA.14

Mostaghimi et al12 studied health care resource utilization and all-cause direct health care costs in privately insured AA patients with or without alopecia totalis (AT) or alopecia universalis (AU)(n=14,972) matched with non-AA controls (n=44,916)(1:3 ratio). Mean total all-cause medical and pharmacy costs were higher in both AA groups compared with controls (AT/AU, $18,988 vs $11,030; non-AT/AU, $13,686 vs $9336; P<.001 for both). Out-of-pocket costs were higher for AA vs controls (AT/AU, $2685 vs $1457; non-AT/AU, $2223 vs $1341; P<.001 for both). Medical costs in the AT/AU and non-AT/AU groups largely were driven by outpatient costs (AT/AU, $10,277 vs $5713; non-AT/AU, $8078 vs $4672; P<.001 for both).12

Costs of Concealment—When studying the out-of-pocket costs of AA (N=675), Li et al13 discovered that the median yearly spending was highest on headwear or cosmetic items such as hats, wigs, and makeup ($450; interquartile range, $50–$1500). Mesinkovska et al14 reported that 49% of patients had insurance that covered AA treatment. However, 75% of patients reported that their insurance would not cover costs of concealment (eg, weave, wig, hair piece). Patients (N=112) spent a mean of $2211 per year and 10.3 hours per week on concealment.14

Minoxidil—Minoxidil solution is available over-the-counter, and its ease of access makes it a popular treatment for AA.17 Because manufacturers can sell directly to the public, minoxidil is marketed with bold claims and convincing packaging. Shrank18 noted that the product can take 4 months to work, meaning customers must incur a substantial cost burden before realizing the treatment’s benefit, which is not always obvious when purchasing minoxidil products, leaving customers—who were marketed a miracle drug—disappointed. Per Shrank,18 patients who did not experience hair regrowth after 4 months were advised to continue treatment for a year, leading them to spend hundreds of dollars for uncertain results. Those who did experience hair regrowth were advised to continue using the product twice daily 7 days per week indefinitely.18

Wehner et al19 studied the association between gender and drug cost for over-the-counter minoxidil. The price that women paid for 2% regular-strength minoxidil solutions was similar to the price that men paid for 5% extra-strength minoxidil solutions (women’s 2%, $7.63/30 mL; men’s 5%, $7.61/30 mL; P=.67). Minoxidil 5% foams with identical ingredients were priced significantly more per volume of the same product when sold as a product directed at women vs a product directed at men (men’s 5%, $8.05/30 mL; women’s 5%, $11.27/30 mL; P<.001).19

 

 

Beach20 compared the cost of oral minoxidil to topical minoxidil. At $28.60 for a 3-month supply, oral minoxidil demonstrated cost savings compared to topical minoxidil ($48.30).20

Diphencyprone—Bhat et al21 studied the cost-efficiency of diphencyprone (DPC) in patients with AA resistant to at least 2 conventional treatments (N=29). After initial sensitization with 2% DPC, patients received weekly or fortnightly treatments. Most of the annual cost burden of DPC treatment was due to staff time and overhead rather than the cost of the DPC itself: $258 for the DPC, $978 in staff time and overhead for the department, and $1233 directly charged to the patient.21

Lekhavat et al22 studied the economic impact of home-use vs office-use DPC in extensive AA (N=82). Both groups received weekly treatments in the hospital until DPC concentrations had been adjusted. Afterward, the home group was given training on self-applying DPC at home. The home group had monthly office visits for DPC concentration evaluation and refills, while the office group had weekly appointments for DPC treatment at the hospital. Calculated costs included those to the health care provider (ie, material, labor, capital costs) and the patient’s final out-of-pocket expense. The total cost to the health care provider was higher for the office group than the home group at 48 weeks (office, $683.52; home, $303.67; P<.001). Median out-of-pocket costs did not vary significantly between groups, which may have been due to small sample size affecting the range (office, $418.07; home, $189.69; P=.101). There was no significant difference between groups in the proportion of patients who responded favorably to the DPC.22

JAK Inhibitors—Chen et al23 studied the efficacy of low-dose (5 mg) tofacitinib to treat severe AA (N=6). Compared to prior studies,24-27 this analysis reported the efficacy of low-dose tofacitinib was not inferior to higher doses (10–20 mg), and low-dose tofacitinib reduced treatment costs by more than 50%.23

Per the GlobalData Healthcare database, the estimated annual cost of therapy for JAK inhibitors following US Food and Drug Administration approval was $50,000. At the time of their reporting, the next most expensive immunomodulatory drug for AA was cyclosporine, with an annual cost of therapy of $1400.28 Dillon29 reviewed the use of JAK inhibitors for the treatment of AA. The cost estimates by Dillon29 prior to FDA approval aligned with the pricing of Eli Lilly and Company for the now-approved JAK inhibitor baricitinib.30 The list price of baricitinib is $2739.99 for a 30-day supply of 2-mg tablets or $5479.98 for a 30-day supply of 4-mg tablets. This amounts to $32,879.88 for an annual supply of 2-mg tablets and $65,759.76 for an annual supply for 4-mg tablets, though the out-of-pocket costs will vary.30

Comment

We reviewed the global and treatment-specific costs of AA, consolidating the available data for the practicing dermatologist. Ten studies of approximately 16,000 patients with AA across a range of levels of evidence (1a to 4) were included (Table). Three of 10 articles studied global costs of AA, 1 studied costs of concealment, 3 studied costs of minoxidil, 2 studied costs of DPC, and 2 studied costs of JAK inhibitors. Only 2 studies achieved level of evidence 1a: the first assessed the economic impact of home-use vs office-use DPC,22 and the second researched the efficacy and outcomes of JAK inhibitors.29

CT113004185_Table_part1.jpg

CT113004185_Table_part2.jpg

Hair-loss treatments and concealment techniques cost the average patient thousands of dollars. Spending was highest on headwear or cosmetic items, which were rarely covered by insurance.13 Psychosocial sequelae further increased cost via therapy charges and lost time at work.14 Patients with AA had greater all-cause medical costs than those without AA, with most of the cost driven by outpatient visits. Patients with AA also paid nearly twice as much as non-AA patients on out-of-pocket health care expenses.14 Despite the high costs and limited efficacy of many AA therapies, patients reported willingness to incur debt or use savings to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress.13

 

 

Minoxidil solution does not require physician office visits and is available over-the-counter.17 Despite identical ingredients, minoxidil is priced more per volume when marketed to women compared with men, which reflects the larger issue of gender-based pricing that does not exist for other AAD-approved alopecia therapies but may exist for cosmetic treatments and nonapproved therapies (eg, vitamins/supplements) that are popular in the treatment of AA.19 Oral minoxidil was more cost-effective than the topical form, and gender-based pricing was a nonissue.20 However, oral minoxidil requires a prescription, mandating patients incur the cost of an office visit. Patients should be wary of gender- or marketing-related surcharges for minoxidil solutions, and oral minoxidil may be a cost-effective choice.

Diphencyprone is a relatively affordable drug for AA, but the regular office visits traditionally required for its administration increase associated cost.21 Self-administration of DPC at home was more cost- and time-effective than in-office DPC administration and did not decrease efficacy. A regimen combining office visits for initial DPC titration, at-home DPC administration, and periodic office follow-up could minimize costs while preserving outcomes and safety.22

Janus kinase inhibitors are cutting-edge and expensive therapies for AA. The annual cost of these medications poses a tremendous burden on the payer (list price of annual supply ritlecitinib is $49,000),31 be that the patient or the insurance company. Low-dose tofacitinib may be similarly efficacious and could substantially reduce treatment costs.23 The true utility of these medications, specifically considering their steep costs, remains to be determined.

Conclusion

Alopecia areata poses a substantial and recurring cost burden on patients that is multifactorial including treatment, office visits, concealment, alternative therapies, psychosocial costs, and missed time at work. Although several treatment options exist, none of them are definitive. Oral minoxidil and at-home DPC administration can be cost-effective, though the cumulative cost is still high. The cost utility of JAK inhibitors remains unclear. When JAK inhibitors are prescribed, low-dose therapy may be used as maintenance to curb treatment costs. Concealment and therapy costs pose an additional, largely out-of-pocket financial burden. Despite the limited efficacy of many AA therapies, patients incur substantial expenses to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress. There are no head-to-head studies comparing the cost-effectiveness of the different AA therapies; thus, it is unclear if one treatment is most efficacious. This topic remains an avenue for future investigation. Much of the cost burden of AA treatment falls directly on patients. Increasing coverage of AA-associated expenses, such as minoxidil therapy or wigs, could decrease the cost burden on patients. Providers also can inform patients about cost-saving tactics, such as purchasing minoxidil based on concentration and vehicle rather than marketing directed at men vs women. Finally, some patients may have insurance plans that at least partially cover the costs of wigs but may not be aware of this benefit. Querying a patient’s insurance provider can further minimize costs.

References
  1. Tosti A, Piraccini BM, Pazzaglia M, et al. Clobetasol propionate 0.05% under occlusion in the treatment of alopecia totalis/universalis. J Am Acad Dermatol. 2003;49:96-98. doi:10.1067/mjd.2003.423
  2. Strazzulla LC, Wang EHC, Avila L, et al. Alopecia areata: an appraisal of new treatment approaches and overview of current therapies. J Am Acad Dermatol. 2018;78:15-24. doi:10.1016/j.jaad.2017.04.1142
  3. Olsen EA, Carson SC, Turney EA. Systemic steroids with or without 2% topical minoxidil in the treatment of alopecia areata. Arch Dermatol. 1992;128:1467-1473.
  4. Levy LL, Urban J, King BA. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. J Am Acad Dermatol. 2015;73:395-399. doi:10.1016/j.jaad.2015.06.045
  5. Ports WC, Khan S, Lan S, et al. A randomized phase 2a efficacy and safety trial of the topical Janus kinase inhibitor tofacitinib in the treatment of chronic plaque psoriasis. Br J Dermatol. 2013;169:137-145. doi:10.1111/bjd.12266
  6. Strober B, Buonanno M, Clark JD, et al. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol. 2013;169:992-999. doi:10.1111/bjd.12517
  7. van der Steen PH, van Baar HM, Happle R, et al. Prognostic factors in the treatment of alopecia areata with diphenylcyclopropenone. J Am Acad Dermatol. 1991;24(2, pt 1):227-230. doi:10.1016/0190-9622(91)70032-w
  8. Strazzulla LC, Avila L, Lo Sicco K, et al. Image gallery: treatment of refractory alopecia universalis with oral tofacitinib citrate and adjunct intralesional triamcinolone injections. Br J Dermatol. 2017;176:E125. doi:10.1111/bjd.15483
  9. Madani S, Shapiro J. Alopecia areata update. J Am Acad Dermatol. 2000;42:549-566; quiz 567-570.
  10. Carnahan MC, Goldstein DA. Ocular complications of topical, peri-ocular, and systemic corticosteroids. Curr Opin Ophthalmol. 2000;11:478-483. doi:10.1097/00055735-200012000-00016
  11. Harel S, Higgins CA, Cerise JE, et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. Sci Adv. 2015;1:E1500973. doi:10.1126/sciadv.1500973
  12. Mostaghimi A, Gandhi K, Done N, et al. All-cause health care resource utilization and costs among adults with alopecia areata: a retrospective claims database study in the United States. J Manag Care Spec Pharm. 2022;28:426-434. doi:10.18553/jmcp.2022.28.4.426
  13. Li SJ, Mostaghimi A, Tkachenko E, et al. Association of out-of-pocket health care costs and financial burden for patients with alopecia areata. JAMA Dermatol. 2019;155:493-494. doi:10.1001/jamadermatol.2018.5218
  14. Mesinkovska N, King B, Mirmirani P, et al. Burden of illness in alopecia areata: a cross-sectional online survey study. J Investig Dermatol Symp Proc. 2020;20:S62-S68. doi:10.1016/j.jisp.2020.05.007
  15. Iuga AO, McGuire MJ. Adherence and health care costs. Risk Manag Healthc Policy. 2014;7:35-44. doi:10.2147/rmhp.S19801
  16. Oxford Centre for Evidence-Based Medicine: Levels of Evidence (March 2009). University of Oxford website. Accessed March 25, 2024. https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009
  17. Klifto KM, Othman S, Kovach SJ. Minoxidil, platelet-rich plasma (PRP), or combined minoxidil and PRP for androgenetic alopecia in men: a cost-effectiveness Markov decision analysis of prospective studies. Cureus. 2021;13:E20839. doi:10.7759/cureus.20839
  18. Shrank AB. Minoxidil over the counter. BMJ. 1995;311:526. doi:10.1136/bmj.311.7004.526
  19. Wehner MR, Nead KT, Lipoff JB. Association between gender and drug cost for over-the-counter minoxidil. JAMA Dermatol. 2017;153:825-826.
  20. Beach RA. Case series of oral minoxidil for androgenetic and traction alopecia: tolerability & the five C’s of oral therapy. Dermatol Ther. 2018;31:E12707. doi:10.1111/dth.12707
  21. Bhat A, Sripathy K, Wahie S, et al. Efficacy and cost-efficiency of diphencyprone for alopecia areata. Br J Dermatol. 2011;165:43-44.
  22. Lekhavat C, Rattanaumpawan P, Juengsamranphong I. Economic impact of home-use versus office-use diphenylcyclopropenone in extensive alopecia areata. Skin Appendage Disord. 2022;8:108-117.
  23. Chen YY, Lin SY, Chen YC, et al. Low-dose tofacitinib for treating patients with severe alopecia areata: an efficient and cost-saving regimen. Eur J Dermatol. 2019;29:667-669. doi:10.1684/ejd.2019.3668
  24. Liu LY, Craiglow BG, Dai F, et al. Tofacitinib for the treatment of severe alopecia areata and variants: a study of 90 patients. J Am Acad Dermatol. 2017;76:22-28. doi:10.1016/j.jaad.2016.09.007
  25. Kennedy Crispin M, Ko JM, Craiglow BG, et al. Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with alopecia areata. JCI Insight. 2016;1:e89776. doi:10.1172/jci.insight.89776
  26. Jabbari A, Sansaricq F, Cerise J, et al. An open-label pilot study to evaluate the efficacy of tofacitinib in moderate to severe patch-type alopecia areata, totalis, and universalis. J Invest Dermatol. 2018;138:1539-1545. doi:10.1016/j.jid.2018.01.032
  27. Craiglow BG, Liu LY, King BA. Tofacitinib for the treatment of alopecia areata and variants in adolescents. J Am Acad Dermatol. 2017;76:29-32. doi:10.1016/j.jaad.2016.09.006
  28. GlobalData Healthcare. Can JAK inhibitors penetrate the alopecia areata market effectively? Pharmaceutical Technology. July 15, 2019. Accessed February 8, 2024. https://www.pharmaceutical-technology.com/analyst-comment/alopecia-areata-treatment-2019/
  29. Dillon KL. A comprehensive literature review of JAK inhibitors in treatment of alopecia areata. Clin Cosmet Investig Dermatol. 2021;14:691-714. doi:10.2147/ccid.S309215
  30. How much should I expect to pay for Olumiant? Accessed March 20, 2024. https://www.lillypricinginfo.com/olumiant
  31. McNamee A. FDA approves first-ever adolescent alopecia treatment from Pfizer. Pharmaceutical Technology. June 26, 2023. Accessed March 20, 2024. https://www.pharmaceutical-technology.com/news/fda-approves-first-ever-adolescent-alopecia-treatment-from-pfizer/?cf-view
References
  1. Tosti A, Piraccini BM, Pazzaglia M, et al. Clobetasol propionate 0.05% under occlusion in the treatment of alopecia totalis/universalis. J Am Acad Dermatol. 2003;49:96-98. doi:10.1067/mjd.2003.423
  2. Strazzulla LC, Wang EHC, Avila L, et al. Alopecia areata: an appraisal of new treatment approaches and overview of current therapies. J Am Acad Dermatol. 2018;78:15-24. doi:10.1016/j.jaad.2017.04.1142
  3. Olsen EA, Carson SC, Turney EA. Systemic steroids with or without 2% topical minoxidil in the treatment of alopecia areata. Arch Dermatol. 1992;128:1467-1473.
  4. Levy LL, Urban J, King BA. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. J Am Acad Dermatol. 2015;73:395-399. doi:10.1016/j.jaad.2015.06.045
  5. Ports WC, Khan S, Lan S, et al. A randomized phase 2a efficacy and safety trial of the topical Janus kinase inhibitor tofacitinib in the treatment of chronic plaque psoriasis. Br J Dermatol. 2013;169:137-145. doi:10.1111/bjd.12266
  6. Strober B, Buonanno M, Clark JD, et al. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. Br J Dermatol. 2013;169:992-999. doi:10.1111/bjd.12517
  7. van der Steen PH, van Baar HM, Happle R, et al. Prognostic factors in the treatment of alopecia areata with diphenylcyclopropenone. J Am Acad Dermatol. 1991;24(2, pt 1):227-230. doi:10.1016/0190-9622(91)70032-w
  8. Strazzulla LC, Avila L, Lo Sicco K, et al. Image gallery: treatment of refractory alopecia universalis with oral tofacitinib citrate and adjunct intralesional triamcinolone injections. Br J Dermatol. 2017;176:E125. doi:10.1111/bjd.15483
  9. Madani S, Shapiro J. Alopecia areata update. J Am Acad Dermatol. 2000;42:549-566; quiz 567-570.
  10. Carnahan MC, Goldstein DA. Ocular complications of topical, peri-ocular, and systemic corticosteroids. Curr Opin Ophthalmol. 2000;11:478-483. doi:10.1097/00055735-200012000-00016
  11. Harel S, Higgins CA, Cerise JE, et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. Sci Adv. 2015;1:E1500973. doi:10.1126/sciadv.1500973
  12. Mostaghimi A, Gandhi K, Done N, et al. All-cause health care resource utilization and costs among adults with alopecia areata: a retrospective claims database study in the United States. J Manag Care Spec Pharm. 2022;28:426-434. doi:10.18553/jmcp.2022.28.4.426
  13. Li SJ, Mostaghimi A, Tkachenko E, et al. Association of out-of-pocket health care costs and financial burden for patients with alopecia areata. JAMA Dermatol. 2019;155:493-494. doi:10.1001/jamadermatol.2018.5218
  14. Mesinkovska N, King B, Mirmirani P, et al. Burden of illness in alopecia areata: a cross-sectional online survey study. J Investig Dermatol Symp Proc. 2020;20:S62-S68. doi:10.1016/j.jisp.2020.05.007
  15. Iuga AO, McGuire MJ. Adherence and health care costs. Risk Manag Healthc Policy. 2014;7:35-44. doi:10.2147/rmhp.S19801
  16. Oxford Centre for Evidence-Based Medicine: Levels of Evidence (March 2009). University of Oxford website. Accessed March 25, 2024. https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009
  17. Klifto KM, Othman S, Kovach SJ. Minoxidil, platelet-rich plasma (PRP), or combined minoxidil and PRP for androgenetic alopecia in men: a cost-effectiveness Markov decision analysis of prospective studies. Cureus. 2021;13:E20839. doi:10.7759/cureus.20839
  18. Shrank AB. Minoxidil over the counter. BMJ. 1995;311:526. doi:10.1136/bmj.311.7004.526
  19. Wehner MR, Nead KT, Lipoff JB. Association between gender and drug cost for over-the-counter minoxidil. JAMA Dermatol. 2017;153:825-826.
  20. Beach RA. Case series of oral minoxidil for androgenetic and traction alopecia: tolerability & the five C’s of oral therapy. Dermatol Ther. 2018;31:E12707. doi:10.1111/dth.12707
  21. Bhat A, Sripathy K, Wahie S, et al. Efficacy and cost-efficiency of diphencyprone for alopecia areata. Br J Dermatol. 2011;165:43-44.
  22. Lekhavat C, Rattanaumpawan P, Juengsamranphong I. Economic impact of home-use versus office-use diphenylcyclopropenone in extensive alopecia areata. Skin Appendage Disord. 2022;8:108-117.
  23. Chen YY, Lin SY, Chen YC, et al. Low-dose tofacitinib for treating patients with severe alopecia areata: an efficient and cost-saving regimen. Eur J Dermatol. 2019;29:667-669. doi:10.1684/ejd.2019.3668
  24. Liu LY, Craiglow BG, Dai F, et al. Tofacitinib for the treatment of severe alopecia areata and variants: a study of 90 patients. J Am Acad Dermatol. 2017;76:22-28. doi:10.1016/j.jaad.2016.09.007
  25. Kennedy Crispin M, Ko JM, Craiglow BG, et al. Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with alopecia areata. JCI Insight. 2016;1:e89776. doi:10.1172/jci.insight.89776
  26. Jabbari A, Sansaricq F, Cerise J, et al. An open-label pilot study to evaluate the efficacy of tofacitinib in moderate to severe patch-type alopecia areata, totalis, and universalis. J Invest Dermatol. 2018;138:1539-1545. doi:10.1016/j.jid.2018.01.032
  27. Craiglow BG, Liu LY, King BA. Tofacitinib for the treatment of alopecia areata and variants in adolescents. J Am Acad Dermatol. 2017;76:29-32. doi:10.1016/j.jaad.2016.09.006
  28. GlobalData Healthcare. Can JAK inhibitors penetrate the alopecia areata market effectively? Pharmaceutical Technology. July 15, 2019. Accessed February 8, 2024. https://www.pharmaceutical-technology.com/analyst-comment/alopecia-areata-treatment-2019/
  29. Dillon KL. A comprehensive literature review of JAK inhibitors in treatment of alopecia areata. Clin Cosmet Investig Dermatol. 2021;14:691-714. doi:10.2147/ccid.S309215
  30. How much should I expect to pay for Olumiant? Accessed March 20, 2024. https://www.lillypricinginfo.com/olumiant
  31. McNamee A. FDA approves first-ever adolescent alopecia treatment from Pfizer. Pharmaceutical Technology. June 26, 2023. Accessed March 20, 2024. https://www.pharmaceutical-technology.com/news/fda-approves-first-ever-adolescent-alopecia-treatment-from-pfizer/?cf-view
Page Number
185-190
Page Number
185-190
Publications
Publications
Topics
Article Type
Display Headline
Evaluating the Cost Burden of Alopecia Areata Treatment: A Comprehensive Review for Dermatologists
Display Headline
Evaluating the Cost Burden of Alopecia Areata Treatment: A Comprehensive Review for Dermatologists
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Patel 0424</fileName> <TBEID>0C02F4A7.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F4A7</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Patel</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240408T075020</firstPublished> <LastPublished>20240408T075020</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240408T075019</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Palak V. Patel, BA, BS; Angelica Coello, BS; Jorge Larrondo, MD</byline> <bylineText>Palak V. Patel, BA, BS; Angelica Coello, BS; Jorge Larrondo, MD; Amy McMichael, MD</bylineText> <bylineFull>Palak V. Patel, BA, BS; Angelica Coello, BS; Jorge Larrondo, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>185-190</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Alopecia areata (AA) affects 4.5 million individuals in the United States, with 66% younger than 30 years.1,2 Inflammation causes hair loss in well-circumscribe</metaDescription> <articlePDF>300910</articlePDF> <teaserImage/> <title>Evaluating the Cost Burden of Alopecia Areata Treatment: A Comprehensive Review for Dermatologists</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>hair</keyword> <keyword> alopecia areata</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">219</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026fd.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Evaluating the Cost Burden of Alopecia Areata Treatment: A Comprehensive Review for Dermatologists</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Alopecia areata (AA) is managed with prolonged medical treatments and cosmetic therapies, whose cost can be burdensome. We sought to identify the costs of AA treatment and consolidate the available data for the practicing dermatologist by performing a PubMed search of articles indexed for MEDLINE. Ten studies including approximately 16,000 patients with AA across a range of Oxford Centre for Evidence-Based Medicine Levels of Evidence were included. Studies showed that despite the limited efficacy of many AA therapies, patients incurred substantial expenses to manage their AA.</p> <p>Alopecia areata (AA) affects 4.5 million individuals in the United States, with 66% younger than 30 years.<sup>1,2</sup> Inflammation causes hair loss in well-circumscribed, nonscarring patches on the body with a predilection for the scalp.<sup>3-6</sup> The disease can devastate a patient’s self-esteem, in turn reducing quality of life.<sup>1,7</sup> Alopecia areata is an autoimmune T-cell–mediated disease in which hair follicles lose their immune privilege.<sup>8-10</sup> Several specific mechanisms in the cytokine interactions between T cells and the hair follicle have been discovered, revealing the Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathway as pivotal in the pathogenesis of the disease and leading to the use of JAK inhibitors for treatment.<sup>11</sup></p> <p>There is no cure for AA, and the condition is managed with prolonged medical treatments and cosmetic therapies.<sup>2</sup> Although some patients may be able to manage the annual cost, the cumulative cost of AA treatment can be burdensome.<sup>12</sup> This cumulative cost may increase if newer, potentially expensive treatments become the standard of care. Patients with AA report dipping into their savings (41.3%) and cutting back on food or clothing expenses (33.9%) to account for the cost of alopecia treatment. Although prior estimates of the annual out-of-pocket cost of AA treatments range from $1354 to $2685, the cost burden of individual therapies is poorly understood.<sup>12-14<br/><br/></sup>Patients who must juggle expensive medical bills with basic living expenses may be lost to follow-up or fall into treatment nonadherence.<sup>15</sup> Other patients’ out-of-pocket costs may be manageable, but the costs to the health care system may compromise care in other ways. We conducted a literature review of the recommended therapies for AA based on American Academy of Dermatology (AAD) guidelines to identify the costs of alopecia treatment and consolidate the available data for the practicing dermatologist. </p> <h3>Methods</h3> <p>We conducted a PubMed search of articles indexed for MEDLINE through September 15, 2022, using the terms <i>alopecia</i> and <i>cost</i> plus one of the treatments (n<span class="body">=</span>21) identified by the AAD<sup>2</sup> for the treatment of AA (Figure). The reference lists of included articles were reviewed to identify other potentially relevant studies. Forty-five articles were identified. </p> <p>Given the dearth of cost research in alopecia and the paucity of large prospective studies, we excluded articles that were not available in their full-text form or were not in English (n<span class="body">=</span>3), articles whose primary study topic was not AA or an expert-approved alopecia treatment (n<span class="body">=</span>15), and articles with no concrete cost data (n<span class="body">=</span>17), which yielded 10 relevant articles that we studied using qualitative analysis. <br/><br/>Due to substantial differences in study methods and outcome measures, we did not compare the costs of alopecia among studies and did not perform statistical analysis. The quality of each study was investigated and assigned a level of evidence per the 2009 criteria from the Centre for Evidence-Based Medicine.<sup>16</sup><br/><br/>All cost data were converted into US dollars ($) using the conversion rate from the time of the original article’s publication. </p> <h3>Results</h3> <p><i>Total and Out-of-pocket Costs of AA—</i>Li et al<sup>13</sup> studied out-of-pocket health care costs for AA patients (N<span class="body">=</span>675). Of these participants, 56.9% said their AA was moderately to seriously financially burdensome, and 41.3% reported using their savings to manage these expenses. Participants reported median out-of-pocket spending of $1354 (interquartile range, $537–$3300) annually. The most common categories of expenses were hair appointments (81.8%) and vitamins/supplements (67.7%).<sup>13</sup></p> <p>Mesinkovska et al<sup>14</sup> studied the qualitative and quantitative financial burdens of moderate to severe AA (N<span class="body">=</span>216). Fifty-seven percent of patients reported the financial impact of AA as moderately to severely burdensome with a willingness to borrow money or use savings to cover out-of-pocket costs. Patients without insurance cited cost as a major barrier to obtaining reatment. In addition to direct treatment-related expenses, AA patients spent a mean of $1961 per year on therapy to cope with the disease’s psychological burden. Lost work hours represented another source of financial burden; 61% of patients were employed, and 45% of them reported missing time from their job because of AA.<sup>14<br/><br/></sup>Mostaghimi et al<sup>12</sup> studied health care resource utilization and all-cause direct health care costs in privately insured AA patients with or without alopecia totalis (AT) or alopecia universalis (AU)(n<span class="body">=</span>14,972) matched with non-AA controls (n<span class="body">=</span>44,916)(1:3 ratio). Mean total all-cause medical and pharmacy costs were higher in both AA groups compared with controls (AT/AU, $18,988 vs $11,030; non-AT/AU, $13,686 vs $9336; <i>P</i><span class="body">&lt;</span>.001 for both). Out-of-pocket costs were higher for AA vs controls (AT/AU, $2685 vs $1457; non-AT/AU, $2223 vs $1341; <i>P</i><span class="body">&lt;</span>.001 for both). Medical costs in the AT/AU and non-AT/AU groups largely were driven by outpatient costs (AT/AU, $10,277 vs $5713; non-AT/AU, $8078 vs $4672; <i>P</i><span class="body">&lt;</span>.001 for both).<sup>12<br/><br/></sup><i>Costs of Concealment—</i>When studying the out-of-pocket costs of AA (N<span class="body">=</span>675), Li et al<sup>13</sup> discovered that the median yearly spending was highest on headwear or cosmetic items such as hats, wigs, and makeup ($450; interquartile range, $50–$1500). Mesinkovska et al<sup>14</sup> reported that 49% of patients had insurance that covered AA treatment. However, 75% of patients reported that their insurance would not cover costs of concealment (eg, weave, wig, hair piece). Patients (N<span class="body">=</span>112) spent a mean of $2211 per year and 10.3 hours per week on concealment.<sup>14<br/><br/></sup><i>Minoxidil—</i>Minoxidil solution is available over-the-counter, and its ease of access makes it a popular treatment for AA.<sup>17</sup> Because manufacturers can sell directly to the public, minoxidil is marketed with bold claims and convincing packaging. Shrank<sup>18</sup> noted that the product can take 4 months to work, meaning customers must incur a substantial cost burden before realizing the treatment’s benefit, which is not always obvious when purchasing minoxidil products, leaving customers—who were marketed a miracle drug—disappointed. Per Shrank,<sup>18</sup> patients who did not experience hair regrowth after 4 months were advised to continue treatment for a year, leading them to spend hundreds of dollars for uncertain results. Those who did experience hair regrowth were advised to continue using the product twice daily 7 days per week indefinitely.<sup>18<br/><br/></sup>Wehner et al<sup>19</sup> studied the association between gender and drug cost for over-the-counter minoxidil. The price that women paid for 2% regular-strength minoxidil solutions was similar to the price that men paid for 5% extra-strength minoxidil solutions (women’s 2%, $7.63/30 mL; men’s 5%, $7.61/30 mL; <i>P</i><span class="body">=</span>.67). Minoxidil 5% foams with identical ingredients were priced significantly more per volume of the same product when sold as a product directed at women vs a product directed at men (men’s 5%, $8.05/30 mL; women’s 5%, $11.27/30 mL; <i>P</i><span class="body">&lt;</span>.001).<sup>19<br/><br/></sup>Beach<sup>20</sup> compared the cost of oral minoxidil to topical minoxidil. At $28.60 for a 3-month supply, oral minoxidil demonstrated cost savings compared to topical minoxidil ($48.30).<sup>20<br/><br/></sup><i>Diphencyprone—</i>Bhat et al<sup>21</sup> studied the cost-efficiency of diphencyprone (DPC) in patients with AA resistant to at least 2 conventional treatments (N<span class="body">=</span>29). After initial sensitization with 2% DPC, patients received weekly or fortnightly treatments. Most of the annual cost burden of DPC treatment was due to staff time and overhead rather than the cost of the DPC itself: $258 for the DPC, $978 in staff time and overhead for the department, and $1233 directly charged to the patient.<sup>21<br/><br/></sup>Lekhavat et al<sup>22</sup> studied the economic impact of home-use vs office-use DPC in extensive AA (N<span class="body">=</span>82). Both groups received weekly treatments in the hospital until DPC concentrations had been adjusted. Afterward, the home group was given training on self-applying DPC at home. The home group had monthly office visits for DPC concentration evaluation and refills, while the office group had weekly appointments for DPC treatment at the hospital. Calculated costs included those to the health care provider (ie, material, labor, capital costs) and the patient’s final out-of-pocket expense. The total cost to the health care provider was higher for the office group than the home group at 48 weeks (office, $683.52; home, $303.67; <i>P</i><span class="body">&lt;</span>.001). Median out-of-pocket costs did not vary significantly between groups, which may have been due to small sample size affecting the range (office, $418.07; home, $189.69; <i>P</i><span class="body">=</span>.101). There was no significant difference between groups in the proportion of patients who responded favorably to the DPC.<sup>22<br/><br/></sup><i>JAK Inhibitors—</i>Chen et al<sup>23</sup> studied the efficacy of low-dose (5 mg) tofacitinib to treat severe AA (N<span class="body">=</span>6). Compared to prior studies,<sup>24-27</sup> this analysis reported the efficacy of low-dose tofacitinib was not inferior to higher doses (10–20 mg), and low-dose tofacitinib reduced treatment costs by more than 50%.<sup>23<br/><br/></sup>Per the GlobalData Healthcare database, the estimated annual cost of therapy for JAK inhibitors following US Food and Drug Administration approval was $50,000. At the time of their reporting, the next most expensive immunomodulatory drug for AA was cyclosporine, with an annual cost of therapy of $1400.<sup>28</sup> Dillon<sup>29</sup> reviewed the use of JAK inhibitors for the treatment of AA. The cost estimates by Dillon<sup>29</sup> prior to FDA approval aligned with the pricing of Eli Lilly and Company for the now-approved JAK inhibitor baricitinib.<sup>30</sup> The list price of baricitinib is $2739.99 for a 30-day supply of 2-mg tablets or $5479.98 for a 30-day supply of 4-mg tablets. This amounts to $32,879.88 for an annual supply of 2-mg tablets and $65,759.76 for an annual supply for 4-mg tablets, though the out-of-pocket costs will vary.<sup>30</sup></p> <h3>Comment</h3> <p>We reviewed the global and treatment-specific costs of AA, consolidating the available data for the practicing dermatologist. Ten studies of approximately 16,000 patients with AA across a range of levels of evidence (1a to 4) were included (Table). Three of 10 articles studied global costs of AA, 1 studied costs of concealment, 3 studied costs of minoxidil, 2 studied costs of DPC, and 2 studied costs of JAK inhibitors. Only 2 studies achieved level of evidence 1a: the first assessed the economic impact of home-use vs office-use DPC,<sup>22</sup> and the second researched the efficacy and outcomes of JAK inhibitors.<sup>29</sup></p> <p>Hair-loss treatments and concealment techniques cost the average patient thousands of dollars. Spending was highest on headwear or cosmetic items, which were rarely covered by insurance.<sup>13</sup> Psychosocial sequelae further increased cost via therapy charges and lost time at work.<sup>14</sup> Patients with AA had greater all-cause medical costs than those without AA, with most of the cost driven by outpatient visits. Patients with AA also paid nearly twice as much as non-AA patients on out-of-pocket health care expenses.<sup>14</sup> Despite the high costs and limited efficacy of many AA therapies, patients reported willingness to incur debt or use savings to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress.<sup>13<br/><br/></sup>Minoxidil solution does not require physician office visits and is available over-the-counter.<sup>17</sup> Despite identical ingredients, minoxidil is priced more per volume when marketed to women compared with men, which reflects the larger issue of gender-based pricing that does not exist for other AAD-approved alopecia therapies but may exist for cosmetic treatments and nonapproved therapies (eg, vitamins/supplements) that are popular in the treatment of AA.<sup>19</sup> Oral minoxidil was more cost-effective than the topical form, and gender-based pricing was a nonissue.<sup>20</sup> However, oral minoxidil requires a prescription, mandating patients incur the cost of an office visit. Patients should be wary of gender- or marketing-related surcharges for minoxidil solutions, and oral minoxidil may be a cost-effective choice. <br/><br/>Diphencyprone is a relatively affordable drug for AA, but the regular office visits traditionally required for its administration increase associated cost.<sup>21</sup> Self-administration of DPC at home was more cost- and time-effective than in-office DPC administration and did not decrease efficacy. A regimen combining office visits for initial DPC titration, at-home DPC administration, and periodic office follow-up could minimize costs while preserving outcomes and safety.<sup>22<br/><br/></sup>Janus kinase inhibitors are cutting-edge and expensive therapies for AA. The annual cost of these medications poses a tremendous burden on the payer (list price of annual supply ritlecitinib is $49,000),<sup>31</sup> be that the patient or the insurance company. Low-dose tofacitinib may be similarly efficacious and could substantially reduce treatment costs.<sup>23</sup> The true utility of these medications, specifically considering their steep costs, remains to be determined. </p> <h3>Conclusion</h3> <p>Alopecia areata poses a substantial and recurring cost burden on patients that is multifactorial including treatment, office visits, concealment, alternative therapies, psychosocial costs, and missed time at work. Although several treatment options exist, none of them are definitive. Oral minoxidil and at-home DPC administration can be cost-effective, though the cumulative cost is still high. The cost utility of JAK inhibitors remains unclear. When JAK inhibitors are prescribed, low-dose therapy may be used as maintenance to curb treatment costs. Concealment and therapy costs pose an additional, largely out-of-pocket financial burden. Despite the limited efficacy of many AA therapies, patients incur substantial expenses to manage their AA. This willingness to pay reflects AA’s impact on quality of life and puts these patients at high risk for financial distress. There are no head-to-head studies comparing the cost-effectiveness of the different AA therapies; thus, it is unclear if one treatment is most efficacious. This topic remains an avenue for future investigation. Much of the cost burden of AA treatment falls directly on patients. Increasing coverage of AA-associated expenses, such as minoxidil therapy or wigs, could decrease the cost burden on patients. Providers also can inform patients about cost-saving tactics, such as purchasing minoxidil based on concentration and vehicle rather than marketing directed at men vs women. Finally, some patients may have insurance plans that at least partially cover the costs of wigs but may not be aware of this benefit. Querying a patient’s insurance provider can further minimize costs. </p> <h2>References</h2> <p class="reference"> 1. Tosti A, Piraccini BM, Pazzaglia M, et al. Clobetasol propionate 0.05% under occlusion in the treatment of alopecia totalis/universalis. <i>J Am Acad Dermatol</i>. 2003;49:96-98. doi:10.1067/mjd.2003.423<br/><br/> 2. Strazzulla LC, Wang EHC, Avila L, et al. Alopecia areata: an appraisal of new treatment approaches and overview of current therapies. <i>J Am Acad Dermatol</i>. 2018;78:15-24. doi:10.1016/j.jaad.2017.04.1142<br/><br/> 3. Olsen EA, Carson SC, Turney EA. Systemic steroids with or without 2% topical minoxidil in the treatment of alopecia areata. <i>Arch Dermatol</i>. 1992;128:1467-1473.<br/><br/> 4. Levy LL, Urban J, King BA. Treatment of recalcitrant atopic dermatitis with the oral Janus kinase inhibitor tofacitinib citrate. <i>J Am Acad Dermatol</i>. 2015;73:395-399. doi:10.1016/j.jaad.2015.06.045<br/><br/> 5. Ports WC, Khan S, Lan S, et al. A randomized phase 2a efficacy and safety trial of the topical Janus kinase inhibitor tofacitinib in the treatment of chronic plaque psoriasis. <i>Br J Dermatol</i>. 2013;169:137-145. doi:10.1111/bjd.12266<br/><br/> 6. Strober B, Buonanno M, Clark JD, et al. Effect of tofacitinib, a Janus kinase inhibitor, on haematological parameters during 12 weeks of psoriasis treatment. <i>Br J Dermatol</i>. 2013;169:992-999. doi:10.1111/bjd.12517<br/><br/> 7. van der Steen PH, van Baar HM, Happle R, et al. Prognostic factors in the treatment of alopecia areata with diphenylcyclopropenone. <i>J Am Acad Dermatol</i>. 1991;24(2, pt 1):227-230. doi:10.1016/0190-9622(91)70032-w<br/><br/> 8. Strazzulla LC, Avila L, Lo Sicco K, et al. Image gallery: treatment of refractory alopecia universalis with oral tofacitinib citrate and adjunct intralesional triamcinolone injections. <i>Br J Dermatol</i>. 2017;176:E125. doi:10.1111/bjd.15483<br/><br/> 9. Madani S, Shapiro J. Alopecia areata update. <i>J Am Acad Dermatol</i>. 2000;42:549-566; quiz 567-570. <br/><br/>10. Carnahan MC, Goldstein DA. Ocular complications of topical, peri-ocular, and systemic corticosteroids. <i>Curr Opin Ophthalmol</i>. 2000;11:478-483. doi:10.1097/00055735-200012000-00016<br/><br/>11. Harel S, Higgins CA, Cerise JE, et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. <i>Sci Adv</i>. 2015;1:E1500973. doi:10.1126/sciadv.1500973<br/><br/>12. Mostaghimi A, Gandhi K, Done N, et al. All-cause health care resource utilization and costs among adults with alopecia areata: a retrospective claims database study in the United States. <i>J Manag Care Spec Pharm</i>. 2022;28:426-434. doi:10.18553/jmcp.2022.28.4.426<br/><br/>13. Li SJ, Mostaghimi A, Tkachenko E, et al. Association of out-of-pocket health care costs and financial burden for patients with alopecia areata. <i>JAMA Dermatol</i>. 2019;155:493-494. doi:10.1001/jamadermatol.2018.521814. Mesinkovska N, King B, Mirmirani P, et al. Burden of illness in alopecia areata: a cross-sectional online survey study. <i>J Investig Dermatol Symp Proc</i>. 2020;20:S62-S68. doi:10.1016/j.jisp.2020.05.007<br/><br/>15. Iuga AO, McGuire MJ. Adherence and health care costs. <i>Risk Manag Healthc Policy</i>. 2014;7:35-44. doi:10.2147/rmhp.S19801<br/><br/>16. Oxford Centre for Evidence-Based Medicine: Levels of Evidence (March 2009). University of Oxford website. Accessed March 25, 2024. https://www.cebm.ox.ac.uk/resources/levels-of-evidence/oxford-centre-for-evidence-based-medicine-levels-of-evidence-march-2009<br/><br/>17. Klifto KM, Othman S, Kovach SJ. Minoxidil, platelet-rich plasma (PRP), or combined minoxidil and PRP for androgenetic alopecia in men: a cost-effectiveness Markov decision analysis of prospective studies. <i>Cureus</i>. 2021;13:E20839. doi:10.7759/cureus.20839<br/><br/>18. Shrank AB. Minoxidil over the counter. <i>BMJ</i>. 1995;311:526. doi:10.1136/bmj.311.7004.526<br/><br/>19. Wehner MR, Nead KT, Lipoff JB. Association between gender and drug cost for over-the-counter minoxidil. <i>JAMA Dermatol</i>. 2017;153:825-826.<br/><br/>20. Beach RA. Case series of oral minoxidil for androgenetic and traction alopecia: tolerability &amp; the five C’s of oral therapy. <i>Dermatol Ther</i>. 2018;31:E12707. doi:10.1111/dth.12707<br/><br/>21. Bhat A, Sripathy K, Wahie S, et al. Efficacy and cost-efficiency of diphencyprone for alopecia areata. <i>Br J Dermatol</i>. 2011;165:43-44. <br/><br/>22. Lekhavat C, Rattanaumpawan P, Juengsamranphong I. Economic impact of home-use versus office-use diphenylcyclopropenone in extensive alopecia areata. <i>Skin Appendage Disord</i>. 2022;8:108-117.<br/><br/>23. Chen YY, Lin SY, Chen YC, et al. Low-dose tofacitinib for treating patients with severe alopecia areata: an efficient and cost-saving regimen. <i>Eur J Dermatol</i>. 2019;29:667-669. doi:10.1684/ejd.2019.3668<br/><br/>24. Liu LY, Craiglow BG, Dai F, et al. Tofacitinib for the treatment of severe alopecia areata and variants: a study of 90 patients. <i>J Am Acad Dermatol</i>. 2017;76:22-28. doi:10.1016/j.jaad.2016.09.007<br/><br/>25. Kennedy Crispin M, Ko JM, Craiglow BG, et al. Safety and efficacy of the JAK inhibitor tofacitinib citrate in patients with alopecia areata. <i>JCI Insight</i>. 2016;1:e89776. doi:10.1172/jci.insight.89776<br/><br/>26. Jabbari A, Sansaricq F, Cerise J, et al. An open-label pilot study to evaluate the efficacy of tofacitinib in moderate to severe patch-type alopecia areata, totalis, and universalis. <i>J Invest Dermatol</i>. 2018;138:1539-1545. doi:10.1016/j.jid.2018.01.032<br/><br/>27. Craiglow BG, Liu LY, King BA. Tofacitinib for the treatment of alopecia areata and variants in adolescents. <i>J Am Acad Dermatol</i>. 2017;76:29-32. doi:10.1016/j.jaad.2016.09.006<br/><br/>28. GlobalData Healthcare. Can JAK inhibitors penetrate the alopecia areata market effectively? <i>Pharmaceutical Technology</i>. July 15, 2019. Accessed February 8, 2024. https://www.pharmaceutical-technology.com/analyst-comment/alopecia-areata-treatment-2019/<br/><br/>29. Dillon KL. A comprehensive literature review of JAK inhibitors in treatment of alopecia areata. <i>Clin Cosmet Investig Dermatol</i>. 2021;14:691-714. doi:10.2147/ccid.S309215 <br/><br/>30. How much should I expect to pay for Olumiant? Accessed March 20, 2024. https://www.lillypricinginfo.com/olumiant<br/><br/>31. McNamee A. FDA approves first-ever adolescent alopecia treatment from Pfizer. <i>Pharmaceutical Technology</i>. June 26, 2023. Accessed March 20, 2024. https://www.pharmaceutical-technology.com/news/fda-approves-first-ever-adolescent-alopecia-treatment-from-pfizer/?cf-view</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Palak V. Patel, Angelica Coello, and Dr. McMichael are from the Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, North Carolina. Dr. Larrondo is from the Department of Dermatology, Clínica Alemana Universidad del Desarrollo, Santiago, Chile. </p> <p class="disclosure">Palak V. Patel, Angelica Coello, and Dr. Larrondo report no conflict of interest. Dr. McMichael has received research, speaking, and/or consulting support from AbbVie; Arcutis Biotherapeutics; Bristol Meyers Squibb; Concert Pharmaceuticals, Inc; Eli Lilly and Company; eResearch Technology, Inc; Galderma; Incyte Corporation; Informa Healthcare; Janssen Pharmaceuticals; Johnson &amp; Johnson; L’Oréal; Pfizer; Procter and Gamble; REVIAN, Inc; Samumed; Sanofi-Regeneron; Sun Pharmaceuticls; and UCB. <br/><br/>Correspondence: Palak V. Patel, BA, BS, 1 Medical Center Blvd, Winston-Salem, NC 27157-1071 (palpatel@wakehealth.edu). <br/><br/><em>Cutis. </em>2024 April;113(4):185-190. doi:10.12788/cutis.0994</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Hair loss treatments and concealment techniques cost the average patient thousands of dollars. Much of this cost burden comes from items not covered by insurance.</li> <li>Providers should be wary of gender- or marketing-related surcharges for minoxidil solutions, and oral minoxidil may be a cost-effective option. </li> <li>Self-administering diphencyprone at home is more cost- and time-effective than in-office diphencyprone administration and does not decrease efficacy.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Hair loss treatments and concealment techniques cost the average patient thousands of dollars. Much of this cost burden comes from items not covered by insurance.
  • Providers should be wary of gender- or marketing-related surcharges for minoxidil solutions, and oral minoxidil may be a cost-effective option.
  • Self-administering diphencyprone at home is more cost- and time-effective than in-office diphencyprone administration and does not decrease efficacy.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026FD.SIG
Disable zoom
Off

Depression As a Potential Contributing Factor in Hidradenitis Suppurativa and Associated Racial Gaps

Article Type
Changed
Wed, 03/06/2024 - 12:12
Display Headline
Depression As a Potential Contributing Factor in Hidradenitis Suppurativa and Associated Racial Gaps

Hidradenitis suppurativa (HS)—a chronic, relapsing, inflammatory disorder involving terminal hair follicles in apocrine gland–rich skin—manifests as tender inflamed nodules that transform into abscesses, sinus tracts, and scarring.1,2 The etiology of HS is multifactorial, encompassing lifestyle, microbiota, hormonal status, and genetic and environmental factors. These factors activate the immune system around the terminal hair follicles and lead to hyperkeratosis of the infundibulum of the hair follicles in intertriginous regions. This progresses to follicular occlusion, stasis, and eventual rupture. Bacterial multiplication within the plugged pilosebaceous units further boosts immune activation. Resident and migrated cells of the innate and adaptive immune system then release proinflammatory cytokines such as tumor necrosis factor, IL-1β, and IL-17, which further enhance immune cell influx and inflammation.3,4 This aberrant immune response propagates the production of deep-seated inflammatory nodules and abscesses.3-8

The estimated prevalence of HS is 1% worldwide.9 It is more prevalent in female and Black patients (0.30%) than White patients (0.09%) and is intermediate in prevalence in the biracial population (0.22%).10 Hidradenitis suppurativa is thought to be associated with lower socioeconomic status (SES). In a retrospective analysis of HS patients (N=375), approximately one-third of patients were Black, had advanced disease, and had a notably lower SES.11 Furthermore, HS has been reported to be associated with systemic inflammation and comorbidities such as morbid obesity (38.3%) and hypertension (39.6%) as well as other metabolic syndrome–related disorders and depression (48.1%).1

Hidradenitis suppurativa may contribute to the risk for depression through its substantial impact on health-related quality of life, which culminates in social withdrawal, unemployment, and suicidal thoughts.12 The high prevalence of depression in individuals with HS1 and its association with systemic inflammation13 increases the likelihood that a common genetic predisposition also may exist between both conditions. Because depression frequently has been discovered as a concomitant diagnosis in patients with HS, we hypothesize that a shared genetic susceptibility also may exist between the 2 disorders. Our study sought to explore data on the co-occurrence of depression with HS, including its demographics and racial data.

Methods

We conducted a PubMed search of articles indexed for MEDLINE as well as Google Scholar using the terms depression and hidradenitis suppurativa to obtain all research articles published from 2000 to 2022. Articles were selected based on relevance to the topic of exploration. English-language articles that directly addressed the epidemiology, etiology, pathophysiology, and co-occurrence of both depression and HS with numerical data were included. Articles were excluded if they did not explore the information of interest on these 2 disorders or did not contain clear statistical data of patients with the 2 concurrent medical conditions.

Results

Twenty-two cross-sectional, prospective, and retrospective studies that fit the search criteria were identified and included in the analysis (eTable).1,14-34 Sixteen (72.7%) studies were cross-sectional, 5 (22.7%) were retrospective, and only 1 (4.5%) was a prospective study. Only 6 of the studies provided racial data,1,14,17,26,28,32 and of them, 4 had predominately White patients,1,14,26,32 whereas the other 2 had predominantly Black patients.17,28

CT113003137_eTable_part1.jpg

CT113003137_eTable_part2.jpg

Hidradenitis suppurativa was found to coexist with depression in all the studies, with a prevalence of 1.2% to 48.1%. There also was a higher prevalence of depression in HS patients than in the control patients without HS. Furthermore, a recent study by Wright and colleagues14 stratified the depression prevalence data by age and found a higher prevalence of depression in adults vs children with HS (30% vs 12%).

Comment

Major depression—a chronic and debilitating illness—is the chief cause of disability globally and in the United States alone and has a global lifetime prevalence of 17%.35 In a study of 388 patients diagnosed with depression and 404 community-matched controls who were observed for 10 years, depressed patients had a two-thirds higher likelihood of developing a serious physical illness than controls. The depression-associated elevated risk for serious physical illness persisted after controlling for confounding variables such as alcohol abuse, smoking, and level of physical activity.36 Studies also have demonstrated that HS is more prevalent in Black individuals10 and in individuals of low SES,37 who are mostly the Black and Hispanic populations that experience the highest burden of racial microaggression38 and disparities in health access and outcomes.39,40 The severity and chronicity of major depressive disorder also is higher in Black patients compared with White patients (57% vs 39%).41 Because major depression and HS are most common among Black patients who experience the highest-burden negative financial and health disparities, there may be a shared genetic disposition to both medical conditions.

 

 

Moreover, the common detrimental lifestyle choices associated with patients with depression and HS also suggest the possibility of a collective genetic susceptibility. Patients with depression also report increased consumption of alcohol, tobacco, and illicit substances; sedentary lifestyle leading to obesity; and poor compliance with prescribed medical treatment.42 Smoking and obesity are known contributors to the pathogenesis of HS, and their modification also is known to positively impact the disease course. In a retrospective single-cohort study, 50% of obese HS patients (n=35) reported a substantial decrease in disease severity after a reduction of more than 15% in body mass index over 2 years following bariatric surgery (n=35).43 Patients with HS also have reported disease remission following extensive weight loss.44 In addition, evidence has supported smoking cessation in improving the disease course of HS.43 Because these detrimental lifestyle choices are prevalent in both patients with HS and those with depression, a co-genetic susceptibility also may exist.

Furthermore, depression is characterized by a persistent inflammatory state,13,45 similar to HS.46 Elevated levels of a variety of inflammatory markers, such as C-reactive protein (CRP), IL-6, and soluble intercellular adhesion molecule 1, have been reported in patients with depression compared with healthy controls.13,45 Further analysis found a positive correlation and a strong association between depression and these inflammatory markers.47 Moreover, adipokines regulate inflammatory responses, and adipokines play a role in the pathogenesis of HS. Adipokine levels such as elevated omentin-1 (a recently identified adipokine) were found to be altered in patients with HS compared with controls.48 Results from clinical studies and meta-analyses of patients with depression also have demonstrated that adipokines are dysregulated in this population,49,50 which may be another potential genetic link between depression and HS.

In addition, genetic susceptibility to depression and HS may be shared because the inflammatory markers that have a strong association with depression also have been found to play an important role in HS treatment and disease severity prediction. In a retrospective cohort study of 404 patients, CRP or IL-6 levels were found to be reliable predictors of HS disease severity, which may explain why anti–tumor necrosis factor antibody regimens such as adalimumab and infliximab have clinically ameliorated disease activity in several cases of HS.51 In a study evaluating these drugs, high baseline levels of high-sensitivity CRP and IL-6 were predictive of patient response to infliximab.52 In a meta-analysis evaluating 20,791 participants, an association was found between concurrent depression and CRP. Furthermore, inflammation measured by high levels of CRP or IL-6 was observed to predict future depression.53 If the same inflammatory markers—CRP and IL-6—both play a major role in the disease activity of depression and HS, then a concurrent genetic predisposition may exist.

Conclusion

Understanding the comorbidities, etiologies, and risk factors for the development and progression of HS is an important step toward improved disease management. Available studies on comorbid depression in HS largely involve White patients, and more studies are needed in patients with skin of color, particularly the Black population, who have the highest prevalence of HS.10 Given the evidence for an association between depression and HS, we suggest a large-scale investigation of this patient population that includes a complete medical history, onset of HS in comparison to the onset of depression, and specific measures of disease progress and lifetime management of depression, which may help to increase knowledge about the role of depression in HS and encourage more research in this area. If shared genetic susceptibility is established, aggressive management of depression in patients at risk for HS may reduce disease incidence and severity as well as the psychological burden on patients.

References
  1. Crowley JJ, Mekkes JR, Zouboulis CC, et al. Association of hidradenitis suppurativa disease severity with increased risk for systemic comorbidities. Br J Dermatol. 2014;171:1561-1565.
  2. Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. Clin Cosmet Investig Dermatol. 2017;10:105-115.
  3. Sabat R, Jemec GBE, Matusiak Ł, et al. Hidradenitis suppurativa. Nat Rev Dis Prim. 2020;6:1-20.
  4. Wolk K, Warszawska K, Hoeflich C, et al. Deficiency of IL-22 contributes to a chronic inflammatory disease: pathogenetic mechanisms in acne inversa. J Immunol. 2011;186:1228-1239.
  5. von Laffert M, Helmbold P, Wohlrab J, et al. Hidradenitis suppurativa (acne inversa): early inflammatory events at terminal follicles and at interfollicular epidermis. Exp Dermatol. 2010;19:533-537.
  6. Van Der Zee HH, De Ruiter L, Van Den Broecke DG, et al. Elevated levels of tumour necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 in hidradenitis suppurativa skin: a rationale for targeting TNF-α and IL-1β. Br J Dermatol. 2011;164:1292-1298.
  7. Schlapbach C, Hänni T, Yawalkar N, et al. Expression of the IL-23/Th17 pathway in lesions of hidradenitis suppurativa. J Am Acad Dermatol. 2011;65:790-798.
  8. Kelly G, Hughes R, McGarry T, et al. Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa. Br J Dermatol. 2015;173:1431-1439.
  9. Jemec GBE, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. J Am Acad Dermatol. 2015;73(5 Suppl 1):S4-S7.
  10. Garg A, Kirby JS, Lavian J, et al. Sex- and age-adjusted population analysis of prevalence estimates for hidradenitis suppurativa in the United States. JAMA Dermatol. 2017;153:760-764.
  11. Soliman YS, Hoffman LK, Guzman AK, et al. African American patients with hidradenitis suppurativa have significant health care disparities: a retrospective study. J Cutan Med Surg. 2019;23:334-336.
  12. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations. J Am Acad Dermatol. 2022;86:1092-1101.
  13. Beatriz Currier M, Nemeroff CB. Inflammation and mood disorders: proinflammatory cytokines and the pathogenesis of depression. Antiinflamm Antiallergy Agents Med Chem. 2012;9:212-220.
  14. Wright S, Strunk A, Garg A. Prevalence of depression among children, adolescents, and adults with hidradenitis suppurativa. J Am Acad Dermatol. 2022;86:55-60.
  15. Sampogna F, Fania L, Mastroeni S, et al. Correlation between depression, quality of life and clinical severity in patients with hidradenitis suppurativa. Acta Derm Venereol. 2020;100:1-6.
  16. Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. Br J Dermatol. 2019;180:774-781.
  17. Senthilnathan A, Kolli SS, Cardwell LA, et al. Depression in hidradenitis suppurativa. Br J Dermatol. 2019;181:1087-1088.
  18. Pavon Blanco A, Turner MA, Petrof G, et al. To what extent do disease severity and illness perceptions explain depression, anxiety and quality of life in hidradenitis suppurativa? Br J Dermatol. 2019;180:338-345.
  19. Butt M, Sisic M, Silva C, et al. The associations of depression and coping methods on health-related quality of life for those with hidradenitis suppurativa. J Am Acad Dermatol. 2019;80:1137-1139.
  20. Calao M, Wilson JL, Spelman L, et al. Hidradenitis suppurativa (HS) prevalence, demographics and management pathways in Australia: a population-based cross-sectional study. PLoS One. 2018;13:e0200683.
  21. Ingram JR, Jenkins-Jones S, Knipe DW, et al. Population-based Clinical Practice Research Datalink study using algorithm modelling to identify the true burden of hidradenitis suppurativa. Br J Dermatol. 2018;178:917-924.
  22. Kimball AB, Sundaram M, Gauthier G, et al. The comorbidity burden of hidradenitis suppurativa in the United States: a claims data analysis. Dermatol Ther (Heidelb). 2018;8:557.
  23. Thorlacius L, Cohen AD, Gislason GH, et al. Increased suicide risk in patients with hidradenitis suppurativa. J Invest Dermatol. 2018;138:52-57.
  24. Tiri H, Jokelainen J, Timonen M, et al. Somatic and psychiatric comorbidities of hidradenitis suppurativa in children and adolescents. J Am Acad Dermatol. 2018;79:514-519.
  25. Huilaja L, Tiri H, Jokelainen J, et al. Patients with hidradenitis suppurativa have a high psychiatric disease burden: a Finnish nationwide registry study. J Invest Dermatol. 2018;138:46-51.
  26. Kirby JS, Butt M, Esmann S, et al. Association of resilience with depression and health-related quality of life for patients with hidradenitis suppurativa. JAMA Dermatol. 2017;153:1263.
  27. Egeberg A, Gislason GH, Hansen PR. Risk of major adverse cardiovascular events and all-cause mortality in patients with hidradenitis suppurativa. JAMA Dermatol. 2016;152:429-434.
  28. Vangipuram R, Vaidya T, Jandarov R, et al. Factors contributing to depression and chronic pain in patients with hidradenitis suppurativa: results from a single-center retrospective review. Dermatology. 2016;232:692-695.
  29. Rayner L, Jackson K, Turner M, et al. Integrated mental health assessment in a tertiary medical dermatology service: feasibility and the prevalence of common mental disorder. Br J Dermatol. 2015;173:201.
  30. Shavit E, Dreiher J, Freud T, et al. Psychiatric comorbidities in 3207 patients with hidradenitis suppurativa [published online June 9, 2014]. J Eur Acad Dermatol Venereol. 2015;29:371-376.
  31. Kurek A, Johanne Peters EM, Sabat R, et al. Depression is a frequent co-morbidity in patients with acne inversa. J Dtsch Dermatol Ges. 2013;11:743-749.
  32. Vazquez BG, Alikhan A, Weaver AL, et al. Incidence of hidradenitis suppurativa and associated factors: a population-based study of Olmsted County, Minnesota. J Invest Dermatol. 2013;133:97.
  33. Onderdijk AJ, Van Der Zee HH, Esmann S, et al. Depression in patients with hidradenitis suppurativa [published online February 20, 2012]. J Eur Acad Dermatol Venereol. 2013;27:473-478.
  34. Matusiak Ł, Bieniek A, Szepietowski JC. Psychophysical aspects of hidradenitis suppurativa. Acta Derm Venereol. 2010;90:264-268.
  35. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617-627.
  36. Holahan CJ, Pahl SA, Cronkite RC, et al. Depression and vulnerability to incident physical illness across 10 years. J Affect Disord. 2009;123:222-229.
  37. Deckers IE, Janse IC, van der Zee HH, et al. Hidradenitis suppurativa (HS) is associated with low socioeconomic status (SES): a cross-sectional reference study. J Am Acad Dermatol. 2016;75:755-759.e1.
  38. Williams MT, Skinta MD, Kanter JW, et al. A qualitative study of microaggressions against African Americans on predominantly White campuses. BMC Psychol. 2020;8:1-13.
  39. Dunlop DD, Song J, Lyons JS, et al. Racial/ethnic differences in rates of depression among preretirement adults. Am J Public Health. 2003;93:1945-1952.
  40. Williams DR, Priest N, Anderson NB. Understanding associations among race, socioeconomic status, and health: patterns and prospects. Health Psychol. 2016;35:407-411.
  41. Williams DR, González HM, Neighbors H, et al. Prevalence and distribution of major depressive disorder in African Americans, Caribbean Blacks, and Non-Hispanic Whites: results from the National Survey of American Life. Arch Gen Psychiatry. 2007;64:305-315.
  42. Druss BG, Bradford DW, Rosenheck RA, et al. Mental disorders and use of cardiovascular procedures after myocardial infarction. JAMA. 2000;283:506-511.
  43. Kromann CB, Deckers IE, Esmann S, et al. Risk factors, clinical course and long-term prognosis in hidradenitis suppurativa: a cross-sectional study. Br J Dermatol. 2014;171:819-824.
  44. Sivanand A, Gulliver WP, Josan CK, et al. Weight loss and dietary interventions for hidradenitis suppurativa: a systematic review. J Cutan Med Surg . 2020;24:64-72.
  45. Raedler TJ. Inflammatory mechanisms in major depressive disorder. Curr Opin Psychiatry. 2011;24:519-525.
  46. Rocha VZ, Libby P. Obesity, inflammation, and atherosclerosis. Nat Rev Cardiol. 2009;6:399-409.
  47. Davidson KW, Schwartz JE, Kirkland SA, et al. Relation of inflammation to depression and incident coronary heart disease (from the Canadian Nova Scotia Health Survey [NSHS95] Prospective Population Study). Am J Cardiol. 2009;103:755-761.
  48. González-López MA, Ocejo-Viñals JG, Mata C, et al. Evaluation of serum omentin-1 and apelin concentrations in patients with hidradenitis suppurativa. Postepy Dermatol Alergol. 2021;38:450-454.
  49. Taylor VH, Macqueen GM. The role of adipokines in understanding the associations between obesity and depression. J Obes. 2010;2010:748048.
  50. Setayesh L, Ebrahimi R, Pooyan S, et al. The possible mediatory role of adipokines in the association between low carbohydrate diet and depressive symptoms among overweight and obese women. PLoS One. 2021;16:e0257275 .
  51. Andriano TM, Benesh G, Babbush KM, et al. Serum inflammatory markers and leukocyte profiles accurately describe hidradenitis suppurativa disease severity. Int J Dermatol. 2022;61:1270-1275.
  52. Montaudié H, Seitz-Polski B, Cornille A, et al. Interleukin 6 and high-sensitivity C-reactive protein are potential predictive markers of response to infliximab in hidradenitis suppurativa. J Am Acad Dermatol. 2017;6:156-158.
  53. Colasanto M, Madigan S, Korczak DJ. Depression and inflammation among children and adolescents: a meta-analysis. J Affect Disord. 2020;277:940-948.
Article PDF
Author and Disclosure Information

Dr. Okwundu is from the University of Washington, Trios Health Family Medicine Residency, Kennewick. Dr. McMichael is from the Department of Dermatology, Wake Forest Baptist Health, Winston-Salem, North Carolina.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Nwanneka Okwundu, DO, University of Washington, Trios Health Family Medicine Residency, 320 W 10th Ave, #202, Kennewick, WA 99336 (Nwannekaok@pcom.edu).

Issue
Cutis - 113(3)
Publications
Topics
Page Number
137-140,E1-E2
Sections
Author and Disclosure Information

Dr. Okwundu is from the University of Washington, Trios Health Family Medicine Residency, Kennewick. Dr. McMichael is from the Department of Dermatology, Wake Forest Baptist Health, Winston-Salem, North Carolina.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Nwanneka Okwundu, DO, University of Washington, Trios Health Family Medicine Residency, 320 W 10th Ave, #202, Kennewick, WA 99336 (Nwannekaok@pcom.edu).

Author and Disclosure Information

Dr. Okwundu is from the University of Washington, Trios Health Family Medicine Residency, Kennewick. Dr. McMichael is from the Department of Dermatology, Wake Forest Baptist Health, Winston-Salem, North Carolina.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Nwanneka Okwundu, DO, University of Washington, Trios Health Family Medicine Residency, 320 W 10th Ave, #202, Kennewick, WA 99336 (Nwannekaok@pcom.edu).

Article PDF
Article PDF

Hidradenitis suppurativa (HS)—a chronic, relapsing, inflammatory disorder involving terminal hair follicles in apocrine gland–rich skin—manifests as tender inflamed nodules that transform into abscesses, sinus tracts, and scarring.1,2 The etiology of HS is multifactorial, encompassing lifestyle, microbiota, hormonal status, and genetic and environmental factors. These factors activate the immune system around the terminal hair follicles and lead to hyperkeratosis of the infundibulum of the hair follicles in intertriginous regions. This progresses to follicular occlusion, stasis, and eventual rupture. Bacterial multiplication within the plugged pilosebaceous units further boosts immune activation. Resident and migrated cells of the innate and adaptive immune system then release proinflammatory cytokines such as tumor necrosis factor, IL-1β, and IL-17, which further enhance immune cell influx and inflammation.3,4 This aberrant immune response propagates the production of deep-seated inflammatory nodules and abscesses.3-8

The estimated prevalence of HS is 1% worldwide.9 It is more prevalent in female and Black patients (0.30%) than White patients (0.09%) and is intermediate in prevalence in the biracial population (0.22%).10 Hidradenitis suppurativa is thought to be associated with lower socioeconomic status (SES). In a retrospective analysis of HS patients (N=375), approximately one-third of patients were Black, had advanced disease, and had a notably lower SES.11 Furthermore, HS has been reported to be associated with systemic inflammation and comorbidities such as morbid obesity (38.3%) and hypertension (39.6%) as well as other metabolic syndrome–related disorders and depression (48.1%).1

Hidradenitis suppurativa may contribute to the risk for depression through its substantial impact on health-related quality of life, which culminates in social withdrawal, unemployment, and suicidal thoughts.12 The high prevalence of depression in individuals with HS1 and its association with systemic inflammation13 increases the likelihood that a common genetic predisposition also may exist between both conditions. Because depression frequently has been discovered as a concomitant diagnosis in patients with HS, we hypothesize that a shared genetic susceptibility also may exist between the 2 disorders. Our study sought to explore data on the co-occurrence of depression with HS, including its demographics and racial data.

Methods

We conducted a PubMed search of articles indexed for MEDLINE as well as Google Scholar using the terms depression and hidradenitis suppurativa to obtain all research articles published from 2000 to 2022. Articles were selected based on relevance to the topic of exploration. English-language articles that directly addressed the epidemiology, etiology, pathophysiology, and co-occurrence of both depression and HS with numerical data were included. Articles were excluded if they did not explore the information of interest on these 2 disorders or did not contain clear statistical data of patients with the 2 concurrent medical conditions.

Results

Twenty-two cross-sectional, prospective, and retrospective studies that fit the search criteria were identified and included in the analysis (eTable).1,14-34 Sixteen (72.7%) studies were cross-sectional, 5 (22.7%) were retrospective, and only 1 (4.5%) was a prospective study. Only 6 of the studies provided racial data,1,14,17,26,28,32 and of them, 4 had predominately White patients,1,14,26,32 whereas the other 2 had predominantly Black patients.17,28

CT113003137_eTable_part1.jpg

CT113003137_eTable_part2.jpg

Hidradenitis suppurativa was found to coexist with depression in all the studies, with a prevalence of 1.2% to 48.1%. There also was a higher prevalence of depression in HS patients than in the control patients without HS. Furthermore, a recent study by Wright and colleagues14 stratified the depression prevalence data by age and found a higher prevalence of depression in adults vs children with HS (30% vs 12%).

Comment

Major depression—a chronic and debilitating illness—is the chief cause of disability globally and in the United States alone and has a global lifetime prevalence of 17%.35 In a study of 388 patients diagnosed with depression and 404 community-matched controls who were observed for 10 years, depressed patients had a two-thirds higher likelihood of developing a serious physical illness than controls. The depression-associated elevated risk for serious physical illness persisted after controlling for confounding variables such as alcohol abuse, smoking, and level of physical activity.36 Studies also have demonstrated that HS is more prevalent in Black individuals10 and in individuals of low SES,37 who are mostly the Black and Hispanic populations that experience the highest burden of racial microaggression38 and disparities in health access and outcomes.39,40 The severity and chronicity of major depressive disorder also is higher in Black patients compared with White patients (57% vs 39%).41 Because major depression and HS are most common among Black patients who experience the highest-burden negative financial and health disparities, there may be a shared genetic disposition to both medical conditions.

 

 

Moreover, the common detrimental lifestyle choices associated with patients with depression and HS also suggest the possibility of a collective genetic susceptibility. Patients with depression also report increased consumption of alcohol, tobacco, and illicit substances; sedentary lifestyle leading to obesity; and poor compliance with prescribed medical treatment.42 Smoking and obesity are known contributors to the pathogenesis of HS, and their modification also is known to positively impact the disease course. In a retrospective single-cohort study, 50% of obese HS patients (n=35) reported a substantial decrease in disease severity after a reduction of more than 15% in body mass index over 2 years following bariatric surgery (n=35).43 Patients with HS also have reported disease remission following extensive weight loss.44 In addition, evidence has supported smoking cessation in improving the disease course of HS.43 Because these detrimental lifestyle choices are prevalent in both patients with HS and those with depression, a co-genetic susceptibility also may exist.

Furthermore, depression is characterized by a persistent inflammatory state,13,45 similar to HS.46 Elevated levels of a variety of inflammatory markers, such as C-reactive protein (CRP), IL-6, and soluble intercellular adhesion molecule 1, have been reported in patients with depression compared with healthy controls.13,45 Further analysis found a positive correlation and a strong association between depression and these inflammatory markers.47 Moreover, adipokines regulate inflammatory responses, and adipokines play a role in the pathogenesis of HS. Adipokine levels such as elevated omentin-1 (a recently identified adipokine) were found to be altered in patients with HS compared with controls.48 Results from clinical studies and meta-analyses of patients with depression also have demonstrated that adipokines are dysregulated in this population,49,50 which may be another potential genetic link between depression and HS.

In addition, genetic susceptibility to depression and HS may be shared because the inflammatory markers that have a strong association with depression also have been found to play an important role in HS treatment and disease severity prediction. In a retrospective cohort study of 404 patients, CRP or IL-6 levels were found to be reliable predictors of HS disease severity, which may explain why anti–tumor necrosis factor antibody regimens such as adalimumab and infliximab have clinically ameliorated disease activity in several cases of HS.51 In a study evaluating these drugs, high baseline levels of high-sensitivity CRP and IL-6 were predictive of patient response to infliximab.52 In a meta-analysis evaluating 20,791 participants, an association was found between concurrent depression and CRP. Furthermore, inflammation measured by high levels of CRP or IL-6 was observed to predict future depression.53 If the same inflammatory markers—CRP and IL-6—both play a major role in the disease activity of depression and HS, then a concurrent genetic predisposition may exist.

Conclusion

Understanding the comorbidities, etiologies, and risk factors for the development and progression of HS is an important step toward improved disease management. Available studies on comorbid depression in HS largely involve White patients, and more studies are needed in patients with skin of color, particularly the Black population, who have the highest prevalence of HS.10 Given the evidence for an association between depression and HS, we suggest a large-scale investigation of this patient population that includes a complete medical history, onset of HS in comparison to the onset of depression, and specific measures of disease progress and lifetime management of depression, which may help to increase knowledge about the role of depression in HS and encourage more research in this area. If shared genetic susceptibility is established, aggressive management of depression in patients at risk for HS may reduce disease incidence and severity as well as the psychological burden on patients.

Hidradenitis suppurativa (HS)—a chronic, relapsing, inflammatory disorder involving terminal hair follicles in apocrine gland–rich skin—manifests as tender inflamed nodules that transform into abscesses, sinus tracts, and scarring.1,2 The etiology of HS is multifactorial, encompassing lifestyle, microbiota, hormonal status, and genetic and environmental factors. These factors activate the immune system around the terminal hair follicles and lead to hyperkeratosis of the infundibulum of the hair follicles in intertriginous regions. This progresses to follicular occlusion, stasis, and eventual rupture. Bacterial multiplication within the plugged pilosebaceous units further boosts immune activation. Resident and migrated cells of the innate and adaptive immune system then release proinflammatory cytokines such as tumor necrosis factor, IL-1β, and IL-17, which further enhance immune cell influx and inflammation.3,4 This aberrant immune response propagates the production of deep-seated inflammatory nodules and abscesses.3-8

The estimated prevalence of HS is 1% worldwide.9 It is more prevalent in female and Black patients (0.30%) than White patients (0.09%) and is intermediate in prevalence in the biracial population (0.22%).10 Hidradenitis suppurativa is thought to be associated with lower socioeconomic status (SES). In a retrospective analysis of HS patients (N=375), approximately one-third of patients were Black, had advanced disease, and had a notably lower SES.11 Furthermore, HS has been reported to be associated with systemic inflammation and comorbidities such as morbid obesity (38.3%) and hypertension (39.6%) as well as other metabolic syndrome–related disorders and depression (48.1%).1

Hidradenitis suppurativa may contribute to the risk for depression through its substantial impact on health-related quality of life, which culminates in social withdrawal, unemployment, and suicidal thoughts.12 The high prevalence of depression in individuals with HS1 and its association with systemic inflammation13 increases the likelihood that a common genetic predisposition also may exist between both conditions. Because depression frequently has been discovered as a concomitant diagnosis in patients with HS, we hypothesize that a shared genetic susceptibility also may exist between the 2 disorders. Our study sought to explore data on the co-occurrence of depression with HS, including its demographics and racial data.

Methods

We conducted a PubMed search of articles indexed for MEDLINE as well as Google Scholar using the terms depression and hidradenitis suppurativa to obtain all research articles published from 2000 to 2022. Articles were selected based on relevance to the topic of exploration. English-language articles that directly addressed the epidemiology, etiology, pathophysiology, and co-occurrence of both depression and HS with numerical data were included. Articles were excluded if they did not explore the information of interest on these 2 disorders or did not contain clear statistical data of patients with the 2 concurrent medical conditions.

Results

Twenty-two cross-sectional, prospective, and retrospective studies that fit the search criteria were identified and included in the analysis (eTable).1,14-34 Sixteen (72.7%) studies were cross-sectional, 5 (22.7%) were retrospective, and only 1 (4.5%) was a prospective study. Only 6 of the studies provided racial data,1,14,17,26,28,32 and of them, 4 had predominately White patients,1,14,26,32 whereas the other 2 had predominantly Black patients.17,28

CT113003137_eTable_part1.jpg

CT113003137_eTable_part2.jpg

Hidradenitis suppurativa was found to coexist with depression in all the studies, with a prevalence of 1.2% to 48.1%. There also was a higher prevalence of depression in HS patients than in the control patients without HS. Furthermore, a recent study by Wright and colleagues14 stratified the depression prevalence data by age and found a higher prevalence of depression in adults vs children with HS (30% vs 12%).

Comment

Major depression—a chronic and debilitating illness—is the chief cause of disability globally and in the United States alone and has a global lifetime prevalence of 17%.35 In a study of 388 patients diagnosed with depression and 404 community-matched controls who were observed for 10 years, depressed patients had a two-thirds higher likelihood of developing a serious physical illness than controls. The depression-associated elevated risk for serious physical illness persisted after controlling for confounding variables such as alcohol abuse, smoking, and level of physical activity.36 Studies also have demonstrated that HS is more prevalent in Black individuals10 and in individuals of low SES,37 who are mostly the Black and Hispanic populations that experience the highest burden of racial microaggression38 and disparities in health access and outcomes.39,40 The severity and chronicity of major depressive disorder also is higher in Black patients compared with White patients (57% vs 39%).41 Because major depression and HS are most common among Black patients who experience the highest-burden negative financial and health disparities, there may be a shared genetic disposition to both medical conditions.

 

 

Moreover, the common detrimental lifestyle choices associated with patients with depression and HS also suggest the possibility of a collective genetic susceptibility. Patients with depression also report increased consumption of alcohol, tobacco, and illicit substances; sedentary lifestyle leading to obesity; and poor compliance with prescribed medical treatment.42 Smoking and obesity are known contributors to the pathogenesis of HS, and their modification also is known to positively impact the disease course. In a retrospective single-cohort study, 50% of obese HS patients (n=35) reported a substantial decrease in disease severity after a reduction of more than 15% in body mass index over 2 years following bariatric surgery (n=35).43 Patients with HS also have reported disease remission following extensive weight loss.44 In addition, evidence has supported smoking cessation in improving the disease course of HS.43 Because these detrimental lifestyle choices are prevalent in both patients with HS and those with depression, a co-genetic susceptibility also may exist.

Furthermore, depression is characterized by a persistent inflammatory state,13,45 similar to HS.46 Elevated levels of a variety of inflammatory markers, such as C-reactive protein (CRP), IL-6, and soluble intercellular adhesion molecule 1, have been reported in patients with depression compared with healthy controls.13,45 Further analysis found a positive correlation and a strong association between depression and these inflammatory markers.47 Moreover, adipokines regulate inflammatory responses, and adipokines play a role in the pathogenesis of HS. Adipokine levels such as elevated omentin-1 (a recently identified adipokine) were found to be altered in patients with HS compared with controls.48 Results from clinical studies and meta-analyses of patients with depression also have demonstrated that adipokines are dysregulated in this population,49,50 which may be another potential genetic link between depression and HS.

In addition, genetic susceptibility to depression and HS may be shared because the inflammatory markers that have a strong association with depression also have been found to play an important role in HS treatment and disease severity prediction. In a retrospective cohort study of 404 patients, CRP or IL-6 levels were found to be reliable predictors of HS disease severity, which may explain why anti–tumor necrosis factor antibody regimens such as adalimumab and infliximab have clinically ameliorated disease activity in several cases of HS.51 In a study evaluating these drugs, high baseline levels of high-sensitivity CRP and IL-6 were predictive of patient response to infliximab.52 In a meta-analysis evaluating 20,791 participants, an association was found between concurrent depression and CRP. Furthermore, inflammation measured by high levels of CRP or IL-6 was observed to predict future depression.53 If the same inflammatory markers—CRP and IL-6—both play a major role in the disease activity of depression and HS, then a concurrent genetic predisposition may exist.

Conclusion

Understanding the comorbidities, etiologies, and risk factors for the development and progression of HS is an important step toward improved disease management. Available studies on comorbid depression in HS largely involve White patients, and more studies are needed in patients with skin of color, particularly the Black population, who have the highest prevalence of HS.10 Given the evidence for an association between depression and HS, we suggest a large-scale investigation of this patient population that includes a complete medical history, onset of HS in comparison to the onset of depression, and specific measures of disease progress and lifetime management of depression, which may help to increase knowledge about the role of depression in HS and encourage more research in this area. If shared genetic susceptibility is established, aggressive management of depression in patients at risk for HS may reduce disease incidence and severity as well as the psychological burden on patients.

References
  1. Crowley JJ, Mekkes JR, Zouboulis CC, et al. Association of hidradenitis suppurativa disease severity with increased risk for systemic comorbidities. Br J Dermatol. 2014;171:1561-1565.
  2. Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. Clin Cosmet Investig Dermatol. 2017;10:105-115.
  3. Sabat R, Jemec GBE, Matusiak Ł, et al. Hidradenitis suppurativa. Nat Rev Dis Prim. 2020;6:1-20.
  4. Wolk K, Warszawska K, Hoeflich C, et al. Deficiency of IL-22 contributes to a chronic inflammatory disease: pathogenetic mechanisms in acne inversa. J Immunol. 2011;186:1228-1239.
  5. von Laffert M, Helmbold P, Wohlrab J, et al. Hidradenitis suppurativa (acne inversa): early inflammatory events at terminal follicles and at interfollicular epidermis. Exp Dermatol. 2010;19:533-537.
  6. Van Der Zee HH, De Ruiter L, Van Den Broecke DG, et al. Elevated levels of tumour necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 in hidradenitis suppurativa skin: a rationale for targeting TNF-α and IL-1β. Br J Dermatol. 2011;164:1292-1298.
  7. Schlapbach C, Hänni T, Yawalkar N, et al. Expression of the IL-23/Th17 pathway in lesions of hidradenitis suppurativa. J Am Acad Dermatol. 2011;65:790-798.
  8. Kelly G, Hughes R, McGarry T, et al. Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa. Br J Dermatol. 2015;173:1431-1439.
  9. Jemec GBE, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. J Am Acad Dermatol. 2015;73(5 Suppl 1):S4-S7.
  10. Garg A, Kirby JS, Lavian J, et al. Sex- and age-adjusted population analysis of prevalence estimates for hidradenitis suppurativa in the United States. JAMA Dermatol. 2017;153:760-764.
  11. Soliman YS, Hoffman LK, Guzman AK, et al. African American patients with hidradenitis suppurativa have significant health care disparities: a retrospective study. J Cutan Med Surg. 2019;23:334-336.
  12. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations. J Am Acad Dermatol. 2022;86:1092-1101.
  13. Beatriz Currier M, Nemeroff CB. Inflammation and mood disorders: proinflammatory cytokines and the pathogenesis of depression. Antiinflamm Antiallergy Agents Med Chem. 2012;9:212-220.
  14. Wright S, Strunk A, Garg A. Prevalence of depression among children, adolescents, and adults with hidradenitis suppurativa. J Am Acad Dermatol. 2022;86:55-60.
  15. Sampogna F, Fania L, Mastroeni S, et al. Correlation between depression, quality of life and clinical severity in patients with hidradenitis suppurativa. Acta Derm Venereol. 2020;100:1-6.
  16. Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. Br J Dermatol. 2019;180:774-781.
  17. Senthilnathan A, Kolli SS, Cardwell LA, et al. Depression in hidradenitis suppurativa. Br J Dermatol. 2019;181:1087-1088.
  18. Pavon Blanco A, Turner MA, Petrof G, et al. To what extent do disease severity and illness perceptions explain depression, anxiety and quality of life in hidradenitis suppurativa? Br J Dermatol. 2019;180:338-345.
  19. Butt M, Sisic M, Silva C, et al. The associations of depression and coping methods on health-related quality of life for those with hidradenitis suppurativa. J Am Acad Dermatol. 2019;80:1137-1139.
  20. Calao M, Wilson JL, Spelman L, et al. Hidradenitis suppurativa (HS) prevalence, demographics and management pathways in Australia: a population-based cross-sectional study. PLoS One. 2018;13:e0200683.
  21. Ingram JR, Jenkins-Jones S, Knipe DW, et al. Population-based Clinical Practice Research Datalink study using algorithm modelling to identify the true burden of hidradenitis suppurativa. Br J Dermatol. 2018;178:917-924.
  22. Kimball AB, Sundaram M, Gauthier G, et al. The comorbidity burden of hidradenitis suppurativa in the United States: a claims data analysis. Dermatol Ther (Heidelb). 2018;8:557.
  23. Thorlacius L, Cohen AD, Gislason GH, et al. Increased suicide risk in patients with hidradenitis suppurativa. J Invest Dermatol. 2018;138:52-57.
  24. Tiri H, Jokelainen J, Timonen M, et al. Somatic and psychiatric comorbidities of hidradenitis suppurativa in children and adolescents. J Am Acad Dermatol. 2018;79:514-519.
  25. Huilaja L, Tiri H, Jokelainen J, et al. Patients with hidradenitis suppurativa have a high psychiatric disease burden: a Finnish nationwide registry study. J Invest Dermatol. 2018;138:46-51.
  26. Kirby JS, Butt M, Esmann S, et al. Association of resilience with depression and health-related quality of life for patients with hidradenitis suppurativa. JAMA Dermatol. 2017;153:1263.
  27. Egeberg A, Gislason GH, Hansen PR. Risk of major adverse cardiovascular events and all-cause mortality in patients with hidradenitis suppurativa. JAMA Dermatol. 2016;152:429-434.
  28. Vangipuram R, Vaidya T, Jandarov R, et al. Factors contributing to depression and chronic pain in patients with hidradenitis suppurativa: results from a single-center retrospective review. Dermatology. 2016;232:692-695.
  29. Rayner L, Jackson K, Turner M, et al. Integrated mental health assessment in a tertiary medical dermatology service: feasibility and the prevalence of common mental disorder. Br J Dermatol. 2015;173:201.
  30. Shavit E, Dreiher J, Freud T, et al. Psychiatric comorbidities in 3207 patients with hidradenitis suppurativa [published online June 9, 2014]. J Eur Acad Dermatol Venereol. 2015;29:371-376.
  31. Kurek A, Johanne Peters EM, Sabat R, et al. Depression is a frequent co-morbidity in patients with acne inversa. J Dtsch Dermatol Ges. 2013;11:743-749.
  32. Vazquez BG, Alikhan A, Weaver AL, et al. Incidence of hidradenitis suppurativa and associated factors: a population-based study of Olmsted County, Minnesota. J Invest Dermatol. 2013;133:97.
  33. Onderdijk AJ, Van Der Zee HH, Esmann S, et al. Depression in patients with hidradenitis suppurativa [published online February 20, 2012]. J Eur Acad Dermatol Venereol. 2013;27:473-478.
  34. Matusiak Ł, Bieniek A, Szepietowski JC. Psychophysical aspects of hidradenitis suppurativa. Acta Derm Venereol. 2010;90:264-268.
  35. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617-627.
  36. Holahan CJ, Pahl SA, Cronkite RC, et al. Depression and vulnerability to incident physical illness across 10 years. J Affect Disord. 2009;123:222-229.
  37. Deckers IE, Janse IC, van der Zee HH, et al. Hidradenitis suppurativa (HS) is associated with low socioeconomic status (SES): a cross-sectional reference study. J Am Acad Dermatol. 2016;75:755-759.e1.
  38. Williams MT, Skinta MD, Kanter JW, et al. A qualitative study of microaggressions against African Americans on predominantly White campuses. BMC Psychol. 2020;8:1-13.
  39. Dunlop DD, Song J, Lyons JS, et al. Racial/ethnic differences in rates of depression among preretirement adults. Am J Public Health. 2003;93:1945-1952.
  40. Williams DR, Priest N, Anderson NB. Understanding associations among race, socioeconomic status, and health: patterns and prospects. Health Psychol. 2016;35:407-411.
  41. Williams DR, González HM, Neighbors H, et al. Prevalence and distribution of major depressive disorder in African Americans, Caribbean Blacks, and Non-Hispanic Whites: results from the National Survey of American Life. Arch Gen Psychiatry. 2007;64:305-315.
  42. Druss BG, Bradford DW, Rosenheck RA, et al. Mental disorders and use of cardiovascular procedures after myocardial infarction. JAMA. 2000;283:506-511.
  43. Kromann CB, Deckers IE, Esmann S, et al. Risk factors, clinical course and long-term prognosis in hidradenitis suppurativa: a cross-sectional study. Br J Dermatol. 2014;171:819-824.
  44. Sivanand A, Gulliver WP, Josan CK, et al. Weight loss and dietary interventions for hidradenitis suppurativa: a systematic review. J Cutan Med Surg . 2020;24:64-72.
  45. Raedler TJ. Inflammatory mechanisms in major depressive disorder. Curr Opin Psychiatry. 2011;24:519-525.
  46. Rocha VZ, Libby P. Obesity, inflammation, and atherosclerosis. Nat Rev Cardiol. 2009;6:399-409.
  47. Davidson KW, Schwartz JE, Kirkland SA, et al. Relation of inflammation to depression and incident coronary heart disease (from the Canadian Nova Scotia Health Survey [NSHS95] Prospective Population Study). Am J Cardiol. 2009;103:755-761.
  48. González-López MA, Ocejo-Viñals JG, Mata C, et al. Evaluation of serum omentin-1 and apelin concentrations in patients with hidradenitis suppurativa. Postepy Dermatol Alergol. 2021;38:450-454.
  49. Taylor VH, Macqueen GM. The role of adipokines in understanding the associations between obesity and depression. J Obes. 2010;2010:748048.
  50. Setayesh L, Ebrahimi R, Pooyan S, et al. The possible mediatory role of adipokines in the association between low carbohydrate diet and depressive symptoms among overweight and obese women. PLoS One. 2021;16:e0257275 .
  51. Andriano TM, Benesh G, Babbush KM, et al. Serum inflammatory markers and leukocyte profiles accurately describe hidradenitis suppurativa disease severity. Int J Dermatol. 2022;61:1270-1275.
  52. Montaudié H, Seitz-Polski B, Cornille A, et al. Interleukin 6 and high-sensitivity C-reactive protein are potential predictive markers of response to infliximab in hidradenitis suppurativa. J Am Acad Dermatol. 2017;6:156-158.
  53. Colasanto M, Madigan S, Korczak DJ. Depression and inflammation among children and adolescents: a meta-analysis. J Affect Disord. 2020;277:940-948.
References
  1. Crowley JJ, Mekkes JR, Zouboulis CC, et al. Association of hidradenitis suppurativa disease severity with increased risk for systemic comorbidities. Br J Dermatol. 2014;171:1561-1565.
  2. Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. Clin Cosmet Investig Dermatol. 2017;10:105-115.
  3. Sabat R, Jemec GBE, Matusiak Ł, et al. Hidradenitis suppurativa. Nat Rev Dis Prim. 2020;6:1-20.
  4. Wolk K, Warszawska K, Hoeflich C, et al. Deficiency of IL-22 contributes to a chronic inflammatory disease: pathogenetic mechanisms in acne inversa. J Immunol. 2011;186:1228-1239.
  5. von Laffert M, Helmbold P, Wohlrab J, et al. Hidradenitis suppurativa (acne inversa): early inflammatory events at terminal follicles and at interfollicular epidermis. Exp Dermatol. 2010;19:533-537.
  6. Van Der Zee HH, De Ruiter L, Van Den Broecke DG, et al. Elevated levels of tumour necrosis factor (TNF)-α, interleukin (IL)-1β and IL-10 in hidradenitis suppurativa skin: a rationale for targeting TNF-α and IL-1β. Br J Dermatol. 2011;164:1292-1298.
  7. Schlapbach C, Hänni T, Yawalkar N, et al. Expression of the IL-23/Th17 pathway in lesions of hidradenitis suppurativa. J Am Acad Dermatol. 2011;65:790-798.
  8. Kelly G, Hughes R, McGarry T, et al. Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa. Br J Dermatol. 2015;173:1431-1439.
  9. Jemec GBE, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. J Am Acad Dermatol. 2015;73(5 Suppl 1):S4-S7.
  10. Garg A, Kirby JS, Lavian J, et al. Sex- and age-adjusted population analysis of prevalence estimates for hidradenitis suppurativa in the United States. JAMA Dermatol. 2017;153:760-764.
  11. Soliman YS, Hoffman LK, Guzman AK, et al. African American patients with hidradenitis suppurativa have significant health care disparities: a retrospective study. J Cutan Med Surg. 2019;23:334-336.
  12. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations. J Am Acad Dermatol. 2022;86:1092-1101.
  13. Beatriz Currier M, Nemeroff CB. Inflammation and mood disorders: proinflammatory cytokines and the pathogenesis of depression. Antiinflamm Antiallergy Agents Med Chem. 2012;9:212-220.
  14. Wright S, Strunk A, Garg A. Prevalence of depression among children, adolescents, and adults with hidradenitis suppurativa. J Am Acad Dermatol. 2022;86:55-60.
  15. Sampogna F, Fania L, Mastroeni S, et al. Correlation between depression, quality of life and clinical severity in patients with hidradenitis suppurativa. Acta Derm Venereol. 2020;100:1-6.
  16. Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. Br J Dermatol. 2019;180:774-781.
  17. Senthilnathan A, Kolli SS, Cardwell LA, et al. Depression in hidradenitis suppurativa. Br J Dermatol. 2019;181:1087-1088.
  18. Pavon Blanco A, Turner MA, Petrof G, et al. To what extent do disease severity and illness perceptions explain depression, anxiety and quality of life in hidradenitis suppurativa? Br J Dermatol. 2019;180:338-345.
  19. Butt M, Sisic M, Silva C, et al. The associations of depression and coping methods on health-related quality of life for those with hidradenitis suppurativa. J Am Acad Dermatol. 2019;80:1137-1139.
  20. Calao M, Wilson JL, Spelman L, et al. Hidradenitis suppurativa (HS) prevalence, demographics and management pathways in Australia: a population-based cross-sectional study. PLoS One. 2018;13:e0200683.
  21. Ingram JR, Jenkins-Jones S, Knipe DW, et al. Population-based Clinical Practice Research Datalink study using algorithm modelling to identify the true burden of hidradenitis suppurativa. Br J Dermatol. 2018;178:917-924.
  22. Kimball AB, Sundaram M, Gauthier G, et al. The comorbidity burden of hidradenitis suppurativa in the United States: a claims data analysis. Dermatol Ther (Heidelb). 2018;8:557.
  23. Thorlacius L, Cohen AD, Gislason GH, et al. Increased suicide risk in patients with hidradenitis suppurativa. J Invest Dermatol. 2018;138:52-57.
  24. Tiri H, Jokelainen J, Timonen M, et al. Somatic and psychiatric comorbidities of hidradenitis suppurativa in children and adolescents. J Am Acad Dermatol. 2018;79:514-519.
  25. Huilaja L, Tiri H, Jokelainen J, et al. Patients with hidradenitis suppurativa have a high psychiatric disease burden: a Finnish nationwide registry study. J Invest Dermatol. 2018;138:46-51.
  26. Kirby JS, Butt M, Esmann S, et al. Association of resilience with depression and health-related quality of life for patients with hidradenitis suppurativa. JAMA Dermatol. 2017;153:1263.
  27. Egeberg A, Gislason GH, Hansen PR. Risk of major adverse cardiovascular events and all-cause mortality in patients with hidradenitis suppurativa. JAMA Dermatol. 2016;152:429-434.
  28. Vangipuram R, Vaidya T, Jandarov R, et al. Factors contributing to depression and chronic pain in patients with hidradenitis suppurativa: results from a single-center retrospective review. Dermatology. 2016;232:692-695.
  29. Rayner L, Jackson K, Turner M, et al. Integrated mental health assessment in a tertiary medical dermatology service: feasibility and the prevalence of common mental disorder. Br J Dermatol. 2015;173:201.
  30. Shavit E, Dreiher J, Freud T, et al. Psychiatric comorbidities in 3207 patients with hidradenitis suppurativa [published online June 9, 2014]. J Eur Acad Dermatol Venereol. 2015;29:371-376.
  31. Kurek A, Johanne Peters EM, Sabat R, et al. Depression is a frequent co-morbidity in patients with acne inversa. J Dtsch Dermatol Ges. 2013;11:743-749.
  32. Vazquez BG, Alikhan A, Weaver AL, et al. Incidence of hidradenitis suppurativa and associated factors: a population-based study of Olmsted County, Minnesota. J Invest Dermatol. 2013;133:97.
  33. Onderdijk AJ, Van Der Zee HH, Esmann S, et al. Depression in patients with hidradenitis suppurativa [published online February 20, 2012]. J Eur Acad Dermatol Venereol. 2013;27:473-478.
  34. Matusiak Ł, Bieniek A, Szepietowski JC. Psychophysical aspects of hidradenitis suppurativa. Acta Derm Venereol. 2010;90:264-268.
  35. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62:617-627.
  36. Holahan CJ, Pahl SA, Cronkite RC, et al. Depression and vulnerability to incident physical illness across 10 years. J Affect Disord. 2009;123:222-229.
  37. Deckers IE, Janse IC, van der Zee HH, et al. Hidradenitis suppurativa (HS) is associated with low socioeconomic status (SES): a cross-sectional reference study. J Am Acad Dermatol. 2016;75:755-759.e1.
  38. Williams MT, Skinta MD, Kanter JW, et al. A qualitative study of microaggressions against African Americans on predominantly White campuses. BMC Psychol. 2020;8:1-13.
  39. Dunlop DD, Song J, Lyons JS, et al. Racial/ethnic differences in rates of depression among preretirement adults. Am J Public Health. 2003;93:1945-1952.
  40. Williams DR, Priest N, Anderson NB. Understanding associations among race, socioeconomic status, and health: patterns and prospects. Health Psychol. 2016;35:407-411.
  41. Williams DR, González HM, Neighbors H, et al. Prevalence and distribution of major depressive disorder in African Americans, Caribbean Blacks, and Non-Hispanic Whites: results from the National Survey of American Life. Arch Gen Psychiatry. 2007;64:305-315.
  42. Druss BG, Bradford DW, Rosenheck RA, et al. Mental disorders and use of cardiovascular procedures after myocardial infarction. JAMA. 2000;283:506-511.
  43. Kromann CB, Deckers IE, Esmann S, et al. Risk factors, clinical course and long-term prognosis in hidradenitis suppurativa: a cross-sectional study. Br J Dermatol. 2014;171:819-824.
  44. Sivanand A, Gulliver WP, Josan CK, et al. Weight loss and dietary interventions for hidradenitis suppurativa: a systematic review. J Cutan Med Surg . 2020;24:64-72.
  45. Raedler TJ. Inflammatory mechanisms in major depressive disorder. Curr Opin Psychiatry. 2011;24:519-525.
  46. Rocha VZ, Libby P. Obesity, inflammation, and atherosclerosis. Nat Rev Cardiol. 2009;6:399-409.
  47. Davidson KW, Schwartz JE, Kirkland SA, et al. Relation of inflammation to depression and incident coronary heart disease (from the Canadian Nova Scotia Health Survey [NSHS95] Prospective Population Study). Am J Cardiol. 2009;103:755-761.
  48. González-López MA, Ocejo-Viñals JG, Mata C, et al. Evaluation of serum omentin-1 and apelin concentrations in patients with hidradenitis suppurativa. Postepy Dermatol Alergol. 2021;38:450-454.
  49. Taylor VH, Macqueen GM. The role of adipokines in understanding the associations between obesity and depression. J Obes. 2010;2010:748048.
  50. Setayesh L, Ebrahimi R, Pooyan S, et al. The possible mediatory role of adipokines in the association between low carbohydrate diet and depressive symptoms among overweight and obese women. PLoS One. 2021;16:e0257275 .
  51. Andriano TM, Benesh G, Babbush KM, et al. Serum inflammatory markers and leukocyte profiles accurately describe hidradenitis suppurativa disease severity. Int J Dermatol. 2022;61:1270-1275.
  52. Montaudié H, Seitz-Polski B, Cornille A, et al. Interleukin 6 and high-sensitivity C-reactive protein are potential predictive markers of response to infliximab in hidradenitis suppurativa. J Am Acad Dermatol. 2017;6:156-158.
  53. Colasanto M, Madigan S, Korczak DJ. Depression and inflammation among children and adolescents: a meta-analysis. J Affect Disord. 2020;277:940-948.
Issue
Cutis - 113(3)
Issue
Cutis - 113(3)
Page Number
137-140,E1-E2
Page Number
137-140,E1-E2
Publications
Publications
Topics
Article Type
Display Headline
Depression As a Potential Contributing Factor in Hidradenitis Suppurativa and Associated Racial Gaps
Display Headline
Depression As a Potential Contributing Factor in Hidradenitis Suppurativa and Associated Racial Gaps
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Okwundu 0324</fileName> <TBEID>0C02F2A0.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F2A0</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Depression As a Potential Contri</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240305T101112</firstPublished> <LastPublished>20240305T101112</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240305T101112</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Nwanneka Okwundu, DO; Amy J. McMichael, MD</byline> <bylineText>Nwanneka Okwundu, DO; Amy J. McMichael, MD </bylineText> <bylineFull>Nwanneka Okwundu, DO; Amy J. McMichael, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>(choose one)</newsDocType> <journalDocType>(choose one)</journalDocType> <linkLabel/> <pageRange>137-140,E1-E2</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Hidradenitis suppurativa (HS)—a chronic, relapsing, inflammatory disorder involving terminal hair follicles in apocrine gland–rich skin—manifests as tender infl</metaDescription> <articlePDF>300459</articlePDF> <teaserImage/> <title>Depression As a Potential Contributing Factor in Hidradenitis Suppurativa and Associated Racial Gaps</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2165</CMSID> </CMSIDs> <keywords> <keyword>psoriasis</keyword> <keyword> skin of color</keyword> <keyword> depression</keyword> <keyword> hidradentitis suppurativa</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Audio | 2165</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> <term>136</term> </sections> <topics> <term canonical="true">281</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026e3.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Depression As a Potential Contributing Factor in Hidradenitis Suppurativa and Associated Racial Gaps</title> <deck/> </itemMeta> <itemContent> <p class="abstract">The etiology of hidradenitis suppurativa (HS)—a chronic, relapsing, inflammatory disorder—is multifactorial, encompassing lifestyle, microbiota, hormonal status, and genetic and environmental factors. These factors propagate the production of deep-seated inflammatory nodules seen in HS through aberrant immune response activation and inflammation. The high prevalence of depression in individuals with HS and its association with systemic inflammation increases the likelihood that depression also may be a contributing etiology to HS. Because depression frequently has been discovered as a concomitant diagnosis in patients with HS, we hypothesize that there is a common susceptibility to depression in patients with HS, which we investigated through a literature search of articles published from 2000 to 2022 involving depression and HS. </p> <p> <em><em>Cutis. </em>2024;113:137-140, E1-E2.</em> </p> <p>Hidradenitis suppurativa (HS)—a chronic, relapsing, inflammatory disorder involving terminal hair follicles in apocrine gland–rich skin—manifests as tender inflamed nodules that transform into abscesses, sinus tracts, and scarring.<sup>1,2</sup> The etiology of HS is multifactorial, encompassing lifestyle, microbiota, hormonal status, and genetic and environmental factors. These factors activate the immune system around the terminal hair follicles and lead to hyperkeratosis of the infundibulum of the hair follicles in intertriginous regions. This progresses to follicular occlusion, stasis, and eventual rupture. Bacterial multiplication within the plugged pilosebaceous units further boosts immune activation. Resident and migrated cells of the innate and adaptive immune system then release proinflammatory cytokines such as tumor necrosis factor, IL-1<span class="body">β</span>, and IL-17, which further enhance immune cell influx and inflammation.<sup>3,4</sup> This aberrant immune response propagates the production of deep-seated inflammatory nodules and abscesses.<sup>3-8</sup> </p> <p>The estimated prevalence of HS is 1% worldwide.<sup>9</sup> It is more prevalent in female and Black patients (0.30%) than White patients (0.09%) and is intermediate in prevalence in the biracial population (0.22%).<sup>10</sup> Hidradenitis suppurativa is thought to be associated with lower socioeconomic status (SES). In a retrospective analysis of HS patients (N<span class="body">=</span>375), approximately one-third of patients were Black, had advanced disease, and had a notably lower SES.<sup>11</sup> Furthermore, HS has been reported to be associated with systemic inflammation and comorbidities such as morbid obesity (38.3%) and hypertension (39.6%) as well as other metabolic syndrome–related disorders and depression (48.1%).<sup>1</sup> <br/><br/>Hidradenitis suppurativa may contribute to the risk for depression through its substantial impact on health-related quality of life, which culminates in social withdrawal, unemployment, and suicidal thoughts.<sup>12</sup> The high prevalence of depression in individuals with HS<sup>1</sup> and its association with systemic inflammation<sup>13</sup> increases the likelihood that a common genetic predisposition also may exist between both conditions. Because depression frequently has been discovered as a concomitant diagnosis in patients with HS, we hypothesize that a shared genetic susceptibility also may exist between the 2 disorders. Our study sought to explore data on the co-occurrence of depression with HS, including its demographics and racial data. </p> <h3>Methods</h3> <p>We conducted a PubMed search of articles indexed for MEDLINE as well as Google Scholar using the terms <i>depression</i> and <i>hidradenitis suppurativa</i> to obtain all research articles published from 2000 to 2022. Articles were selected based on relevance to the topic of exploration. English-language articles that directly addressed the epidemiology, etiology, pathophysiology, and co-occurrence of both depression and HS with numerical data were included. Articles were excluded if they did not explore the information of interest on these 2 disorders or did not contain clear statistical data of patients with the 2 concurrent medical conditions. </p> <h3>Results</h3> <p>Twenty-two cross-sectional, prospective, and retrospective studies that fit the search criteria were identified and included in the analysis (eTable).<sup>1,14-34</sup> Sixteen (72.7%) studies were cross-sectional, 5 (22.7%) were retrospective, and only 1 (4.5%) was a prospective study. Only 6 of the studies provided racial data,<sup>1,14,17,26,28,32</sup> and of them, 4 had predominately White patients,<sup>1,14,26,32</sup> whereas the other 2 had predominantly Black patients.<sup>17,28</sup> </p> <p>Hidradenitis suppurativa was found to coexist with depression in all the studies, with a prevalence of 1.2% to 48.1%. There also was a higher prevalence of depression in HS patients than in the control patients without HS. Furthermore, a recent study by Wright and colleagues<sup>14</sup> stratified the depression prevalence data by age and found a higher prevalence of depression in adults vs children with HS (30% vs 12%).</p> <h3>Comment</h3> <p>Major depression—a chronic and debilitating illness—is the chief cause of disability globally and in the United States alone and has a global lifetime prevalence of 17%.<sup>35</sup> In a study of 388 patients diagnosed with depression and 404 community-matched controls who were observed for 10 years, depressed patients had a two-thirds higher likelihood of developing a serious physical illness than controls. The depression-associated elevated risk for serious physical illness persisted after controlling for confounding variables such as alcohol abuse, smoking, and level of physical activity.<sup>36</sup> Studies also have demonstrated that HS is more prevalent in Black individuals<sup>10</sup> and in individuals of low SES,<sup>37</sup> who are mostly the Black and Hispanic populations that experience the highest burden of racial microaggression<sup>38</sup> and disparities in health access and outcomes.<sup>39,40</sup> The severity and chronicity of major depressive disorder also is higher in Black patients compared with White patients (57% vs 39%).<sup>41</sup> Because major depression and HS are most common among Black patients who experience the highest-burden negative financial and health disparities, there may be a shared genetic disposition to both medical conditions.</p> <p>Moreover, the common detrimental lifestyle choices associated with patients with depression and HS also suggest the possibility of a collective genetic susceptibility. Patients with depression also report increased consumption of alcohol, tobacco, and illicit substances; sedentary lifestyle leading to obesity; and poor compliance with prescribed medical treatment.<sup>42</sup> Smoking and obesity are known contributors to the pathogenesis of HS, and their modification also is known to positively impact the disease course. In a retrospective single-cohort study, 50% of obese HS patients (n<span class="body">=</span>35) reported a substantial decrease in disease severity after a reduction of more than 15% in body mass index over 2 years following bariatric surgery (n<span class="body">=</span>35).<sup>43</sup> Patients with HS also have reported disease remission following extensive weight loss.<sup>44</sup> In addition, evidence has supported smoking cessation in improving the disease course of HS.<sup>43</sup> Because these detrimental lifestyle choices are prevalent in both patients with HS and those with depression, a co-genetic susceptibility also may exist.<br/><br/>Furthermore, depression is characterized by a persistent inflammatory state,<sup>13,45</sup> similar to HS.<sup>46</sup> Elevated levels of a variety of inflammatory markers, such as C-reactive protein (CRP), IL-6, and soluble intercellular adhesion molecule 1, have been reported in patients with depression compared with healthy controls.<sup>13,45</sup> Further analysis found a positive correlation and a strong association between depression and these inflammatory markers.<sup>47</sup> Moreover, adipokines regulate inflammatory responses, and adipokines play a role in the pathogenesis of HS. Adipokine levels such as elevated omentin-1 (a recently identified adipokine) were found to be altered in patients with HS compared with controls.<sup>48</sup> Results from clinical studies and meta-analyses of patients with depression also have demonstrated that adipokines are dysregulated in this population,<sup>49,50</sup> which may be another potential genetic link between depression and HS.<br/><br/>In addition, genetic susceptibility to depression and HS may be shared because the inflammatory markers that have a strong association with depression also have been found to play an important role in HS treatment and disease severity prediction. In a retrospective cohort study of 404 patients, CRP or IL-6 levels were found to be reliable predictors of HS disease severity, which may explain why anti–tumor necrosis factor antibody regimens such as adalimumab and infliximab have clinically ameliorated disease activity in several cases of HS.<sup>51</sup> In a study evaluating these drugs, high baseline levels of high-sensitivity CRP and IL-6 were predictive of patient response to infliximab.<sup>52</sup> In a meta-analysis evaluating 20,791 participants, an association was found between concurrent depression and CRP. Furthermore, inflammation measured by high levels of CRP or IL-6 was observed to predict future depression.<sup>53</sup> If the same inflammatory markers—CRP and IL-6—both play a major role in the disease activity of depression and HS, then a concurrent genetic predisposition may exist.</p> <h3>Conclusion</h3> <p>Understanding the comorbidities, etiologies, and risk factors for the development and progression of HS is an important step toward improved disease management. Available studies on comorbid depression in HS largely involve White patients, and more studies are needed in patients with skin of color, particularly the Black population, who have the highest prevalence of HS.<sup>10</sup> Given the evidence for an association between depression and HS, we suggest a large-scale investigation of this patient population that includes a complete medical history, onset of HS in comparison to the onset of depression, and specific measures of disease progress and lifetime management of depression, which may help to increase knowledge about the role of depression in HS and encourage more research in this area. If shared genetic susceptibility is established, aggressive management of depression in patients at risk for HS may reduce disease incidence and severity as well as the psychological burden on patients.</p> <h2>References</h2> <p class="reference"> 1. Crowley JJ, Mekkes JR, Zouboulis CC, et al. Association of hidradenitis suppurativa disease severity with increased risk for systemic comorbidities. <i>Br J Dermatol</i>. 2014;171:1561-1565. </p> <p class="reference"> 2. Napolitano M, Megna M, Timoshchuk EA, et al. Hidradenitis suppurativa: from pathogenesis to diagnosis and treatment. <i>Clin Cosmet Investig Dermatol</i>. 2017;10:105-115. <br/><br/> 3. Sabat R, Jemec GBE, Matusiak Ł, et al. Hidradenitis suppurativa. <i>Nat Rev Dis Prim</i>. 2020;6:1-20. <br/><br/> 4. Wolk K, Warszawska K, Hoeflich C, et al. Deficiency of IL-22 contributes to a chronic inflammatory disease: pathogenetic mechanisms in acne inversa. <i>J Immunol</i>. 2011;186:1228-1239. <br/><br/> 5. von Laffert M, Helmbold P, Wohlrab J, et al. Hidradenitis suppurativa (acne inversa): early inflammatory events at terminal follicles and at interfollicular epidermis. <i>Exp Dermatol</i>. 2010;19:533-537. <br/><br/> 6. Van Der Zee HH, De Ruiter L, Van Den Broecke DG, et al. Elevated levels of tumour necrosis factor (TNF)-<span class="body">α</span>, interleukin (IL)-1<span class="body">β</span> and IL-10 in hidradenitis suppurativa skin: a rationale for targeting TNF-<span class="body">α</span> and IL-1<span class="body">β</span>. <i>Br J Dermatol</i>. 2011;164:1292-1298.</p> <p class="reference"> 7. Schlapbach C, Hänni T, Yawalkar N, et al. Expression of the IL-23/Th17 pathway in lesions of hidradenitis suppurativa. <i>J Am Acad Dermatol</i>. 2011;65:790-798. <br/><br/> 8. Kelly G, Hughes R, McGarry T, et al. Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa. <i>Br J Dermatol</i>. 2015;173:1431-1439. <br/><br/> 9. Jemec GBE, Kimball AB. Hidradenitis suppurativa: epidemiology and scope of the problem. <i>J Am Acad Dermatol</i>. 2015;73(5 Suppl 1):S4-S7. <br/><br/>10. Garg A, Kirby JS, Lavian J, et al. Sex- and age-adjusted population analysis of prevalence estimates for hidradenitis suppurativa in the United States. <i>JAMA</i> <i>Dermatol</i>. 2017;153:760-764. <br/><br/>11. Soliman YS, Hoffman LK, Guzman AK, et al. African American patients with hidradenitis suppurativa have significant health care disparities: a retrospective study. <i>J Cutan Med Surg</i>. 2019;23:334-336. <br/><br/>12. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations. <i>J Am Acad Dermatol</i>. 2022;86:1092-1101. <br/><br/>13. Beatriz Currier M, Nemeroff CB. Inflammation and mood disorders: proinflammatory cytokines and the pathogenesis of depression. <i>Antiinflamm Antiallergy Agents Med Chem</i>. 2012;9:212-220. <br/><br/>14. Wright S, Strunk A, Garg A. Prevalence of depression among children, adolescents, and adults with hidradenitis suppurativa. <i>J Am Acad Dermatol</i>. 2022;86:55-60. <br/><br/>15. Sampogna F, Fania L, Mastroeni S, et al. Correlation between depression, quality of life and clinical severity in patients with hidradenitis suppurativa. <i>Acta Derm Venereol</i>. 2020;100:1-6. <br/><br/>16. Theut Riis P, Pedersen OB, Sigsgaard V, et al. Prevalence of patients with self-reported hidradenitis suppurativa in a cohort of Danish blood donors: a cross-sectional study. <i>Br J Dermatol</i>. 2019;180:774-781. <br/><br/>17. Senthilnathan A, Kolli SS, Cardwell LA, et al. Depression in hidradenitis suppurativa. <i>Br J Dermatol</i>. 2019;181:1087-1088. <br/><br/>18. Pavon Blanco A, Turner MA, Petrof G, et al. To what extent do disease severity and illness perceptions explain depression, anxiety and quality of life in hidradenitis suppurativa? <i>Br J Dermatol</i>. 2019;180:338-345. <br/><br/>19. Butt M, Sisic M, Silva C, et al. The associations of depression and coping methods on health-related quality of life for those with hidradenitis suppurativa. <i>J Am Acad Dermatol</i>. 2019;80:1137-1139. <br/><br/>20. Calao M, Wilson JL, Spelman L, et al. Hidradenitis suppurativa (HS) prevalence, demographics and management pathways in Australia: a population-based cross-sectional study. <i>PLoS One</i>. 2018;13:e0200683. <br/><br/>21. Ingram JR, Jenkins-Jones S, Knipe DW, et al. Population-based Clinical Practice Research Datalink study using algorithm modelling to identify the true burden of hidradenitis suppurativa. <i>Br J Dermatol</i>. 2018;178:917-924. <br/><br/>22. Kimball AB, Sundaram M, Gauthier G, et al. The comorbidity burden of hidradenitis suppurativa in the United States: a claims data analysis. <i>Dermatol Ther (Heidelb)</i>. 2018;8:557. <br/><br/>23. Thorlacius L, Cohen AD, Gislason GH, et al. Increased suicide risk in patients with hidradenitis suppurativa. <i>J Invest Dermatol</i>. 2018;138:52-57. <br/><br/>24. Tiri H, Jokelainen J, Timonen M, et al. Somatic and psychiatric comorbidities of hidradenitis suppurativa in children and adolescents. <i>J Am Acad Dermatol</i>. 2018;79:514-519. <br/><br/>25. Huilaja L, Tiri H, Jokelainen J, et al. Patients with hidradenitis suppurativa have a high psychiatric disease burden: a Finnish nationwide registry study. <i>J Invest Dermatol</i>. 2018;138:46-51. <br/><br/>26. Kirby JS, Butt M, Esmann S, et al. Association of resilience with depression and health-related quality of life for patients with hidradenitis suppurativa. <i>JAMA Dermatol</i>. 2017;153:1263. <br/><br/>27. Egeberg A, Gislason GH, Hansen PR. Risk of major adverse cardiovascular events and all-cause mortality in patients with hidradenitis suppurativa. <i>JAMA Dermatol</i>. 2016;152:429-434. <br/><br/>28. Vangipuram R, Vaidya T, Jandarov R, et al. Factors contributing to depression and chronic pain in patients with hidradenitis suppurativa: results from a single-center retrospective review. <i>Dermatology.</i> 2016;232:692-695.<br/><br/>29. Rayner L, Jackson K, Turner M, et al. Integrated mental health assessment in a tertiary medical dermatology service: feasibility and the prevalence of common mental disorder. <i>Br J Dermatol</i>. 2015;173:201.<br/><br/>30. Shavit E, Dreiher J, Freud T, et al. Psychiatric comorbidities in 3207 patients with hidradenitis suppurativa [published online June 9, 2014]. <i>J Eur Acad Dermatol Venereol</i>. 2015;29:371-376. <br/><br/>31. Kurek A, Johanne Peters EM, Sabat R, et al. Depression is a frequent co-morbidity in patients with acne inversa. <i>J Dtsch Dermatol Ges</i>. 2013;11:743-749. <br/><br/>32. Vazquez BG, Alikhan A, Weaver AL, et al. Incidence of hidradenitis suppurativa and associated factors: a population-based study of Olmsted County, Minnesota. <i>J Invest Dermatol</i>. 2013;133:97. <br/><br/>33. Onderdijk AJ, Van Der Zee HH, Esmann S, et al. Depression in patients with hidradenitis suppurativa [published online February 20, 2012]. <i>J Eur Acad Dermatol Venereol</i>. 2013;27:473-478. <br/><br/>34. Matusiak Ł, Bieniek A, Szepietowski JC. Psychophysical aspects of hidradenitis suppurativa. <i>Acta Derm Venereol</i>. 2010;90:264-268. <br/><br/>35. Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. <i>Arch Gen Psychiatry</i>. 2005;62:617-627. <br/><br/>36. Holahan CJ, Pahl SA, Cronkite RC, et al. Depression and vulnerability to incident physical illness across 10 years. <i>J Affect Disord</i>. 2009;123:222-229. <br/><br/>37. Deckers IE, Janse IC, van der Zee HH, et al. Hidradenitis suppurativa (HS) is associated with low socioeconomic status (SES): a cross-sectional reference study. <i>J Am Acad Dermatol</i>. 2016;75:755-759.e1. <br/><br/>38. Williams MT, Skinta MD, Kanter JW, et al. A qualitative study of microaggressions against African Americans on predominantly White campuses. <i>BMC Psychol</i>. 2020;8:1-13. </p> <p class="reference">39. Dunlop DD, Song J, Lyons JS, et al. Racial/ethnic differences in rates of depression among preretirement adults. <i>Am J Public Health</i>. 2003;93:1945-1952. <br/><br/>40. Williams DR, Priest N, Anderson NB. Understanding associations among race, socioeconomic status, and health: patterns and prospects. <i>Health Psychol</i>. 2016;35:407-411. <br/><br/>41. Williams DR, González HM, Neighbors H, et al. Prevalence and distribution of major depressive disorder in African Americans, Caribbean Blacks, and Non-Hispanic Whites: results from the National Survey of American Life. <i>Arch Gen Psychiatry</i>. 2007;64:305-315. <br/><br/>42. Druss BG, Bradford DW, Rosenheck RA, et al. Mental disorders and use of cardiovascular procedures after myocardial infarction. <i>JAMA</i>. 2000;283:506-511. <br/><br/>43. Kromann CB, Deckers IE, Esmann S, et al. Risk factors, clinical course and long-term prognosis in hidradenitis suppurativa: a cross-sectional study. <i>Br J Dermatol</i>. 2014;171:819-824. <br/><br/>44. Sivanand A, Gulliver WP, Josan CK, et al. Weight loss and dietary interventions for hidradenitis suppurativa: a systematic review. <i>J Cutan Med Surg</i> . 2020;24:64-72. <br/><br/>45. Raedler TJ. Inflammatory mechanisms in major depressive disorder. <i>Curr Opin Psychiatry</i>. 2011;24:519-525.<br/><br/>46. Rocha VZ, Libby P. Obesity, inflammation, and atherosclerosis. <i>Nat Rev Cardiol</i>. 2009;6:399-409. <br/><br/>47. Davidson KW, Schwartz JE, Kirkland SA, et al. Relation of inflammation to depression and incident coronary heart disease (from the Canadian Nova Scotia Health Survey [NSHS95] Prospective Population Study). <i>Am J Cardiol</i>. 2009;103:755-761. <br/><br/>48. González-López MA, Ocejo-Viñals JG, Mata C, et al. Evaluation of serum omentin-1 and apelin concentrations in patients with hidradenitis suppurativa. <i>Postepy Dermatol Alergol</i>. 2021;38:450-454.<br/><br/>49. Taylor VH, Macqueen GM. The role of adipokines in understanding the associations between obesity and depression. <i>J Obes</i>. 2010;2010:748048. <br/><br/>50. Setayesh L, Ebrahimi R, Pooyan S, et al. The possible mediatory role of adipokines in the association between low carbohydrate diet and depressive symptoms among overweight and obese women. <i>PLoS One</i>. 2021;16:e0257275 . <br/><br/>51. Andriano TM, Benesh G, Babbush KM, et al. Serum inflammatory markers and leukocyte profiles accurately describe hidradenitis suppurativa disease severity. <i>Int J Dermatol</i>. 2022;61:1270-1275. <br/><br/>52. Montaudié H, Seitz-Polski B, Cornille A, et al. Interleukin 6 and high-sensitivity C-reactive protein are potential predictive markers of response to infliximab in hidradenitis suppurativa. <i>J Am Acad Dermatol</i>. 2017;6:156-158. <br/><br/>53. Colasanto M, Madigan S, Korczak DJ. Depression and inflammation among children and adolescents: a meta-analysis. <i>J Affect Disord</i>. 2020;277:940-948. </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. Okwundu is from the University of Washington, Trios Health Family Medicine Residency, Kennewick. Dr. McMichael is from the Department of Dermatology, Wake Forest Baptist Health, Winston-Salem, North Carolina.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>The eTable is available in the Appendix online at www.mdedge.com/dermatology.<br/><br/>Correspondence: Nwanneka Okwundu, DO, University of Washington, Trios Health Family Medicine Residency, 320 W 10th Ave, #202, Kennewick, WA 99336 (Nwannekaok@pcom.edu).doi:10.12788/cutis.0963</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Hidradenitis suppurativa (HS) is known to be associated with systemic inflammation and comorbidities, including depression.</li> <li>Depression may be a potential contributing factor to HS in affected patients, and studies on HS with comorbid depression in patients with skin of color are lacking. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Hidradenitis suppurativa (HS) is known to be associated with systemic inflammation and comorbidities, including depression.
  • Depression may be a potential contributing factor to HS in affected patients, and studies on HS with comorbid depression in patients with skin of color are lacking.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026E3.SIG
Disable zoom
Off

Impact of Ketogenic and Low-Glycemic Diets on Inflammatory Skin Conditions

Article Type
Changed
Tue, 04/23/2024 - 11:50
Display Headline
Impact of Ketogenic and Low-Glycemic Diets on Inflammatory Skin Conditions

Inflammatory skin conditions often have a relapsing and remitting course and represent a large proportion of chronic skin diseases. Common inflammatory skin disorders include acne, psoriasis, hidradenitis suppurativa (HS), atopic dermatitis (AD), and seborrheic dermatitis (SD).1 Although each of these conditions has a unique pathogenesis, they all are driven by a background of chronic inflammation. It has been reported that diets with high levels of refined carbohydrates and saturated or trans-fatty acids may exacerbate existing inflammation.2 Consequently, dietary interventions, such as the ketogenic and low-glycemic diets, have potential anti-inflammatory and metabolic effects that are being assessed as stand-alone or adjunctive therapies for dermatologic diseases.

Diet may partially influence systemic inflammation through its effect on weight. Higher body mass index and obesity are linked to a low-grade inflammatory state and higher levels of circulating inflammatory markers. Therefore, weight loss leads to decreases in inflammatory cytokines, including C-reactive protein, tumor necrosis factor α, and IL-6.3 These cytokines and metabolic effects overlap with inflammatory skin condition pathways. It also is posited that decreased insulin release associated with weight loss results in decreased sebaceous lipogenesis and androgens, which drive keratinocyte proliferation and acne development.4,5 For instance, in a 2015 meta-analysis of 5 randomized controlled trials on psoriasis, patients in the weight loss intervention group had more substantial reductions in psoriasis area and severity index (PASI) scores compared with controls receiving usual care (P=.004).6 However, in a systematic review of 35 studies on acne vulgaris, overweight and obese patients (defined by a body mass index of ≥23 kg/m2) had similar odds of having acne compared with normal-weight individuals (P=.671).7

Similar to weight loss, ketogenesis acts as a negative feedback mechanism to reduce insulin release, leading to decreased inflammation and androgens that often exacerbate inflammatory skin diseases.8 Ketogenesis ensues when daily carbohydrate intake is limited to less than 50 g, and long-term adherence to a ketogenic diet results in metabolic reliance on ketone bodies such as acetoacetate, β-hydroxybutyrate, and acetone.9 These metabolites may decrease free radical damage and consequently improve signs and symptoms of acne, psoriasis, and other inflammatory skin diseases.10-12 Similarly, increased ketones also may decrease activation of the NLRP3 (NOD-, LRR-, and Pyrin domain-containing protein 3) inflammasome and therefore reduce inflammatory markers such as IL-1β and IL-1.4,13 Several proposed mechanisms are outlined in the Table.

CT11302075_Table.jpg

Collectively, low-glycemic and ketogenic diets have been proposed as potential interventions for reducing inflammatory skin conditions. These dietary approaches are hypothesized to exert their effects by facilitating weight loss, elevating ketone levels, and reducing systemic inflammation. The current review summarizes the existing evidence on ketogenic and low-glycemic diets as treatments for inflammatory skin conditions and evaluates the potential benefits of these dietary interventions in managing and improving outcomes for individuals with inflammatory skin conditions.

Methods

Using PubMed for articles indexed for MEDLINE and Google Scholar, a review of the literature was conducted with a combination of the following search terms: low-glycemic diet, inflammatory, dermatologic, ketogenic diet, inflammation, dermatology, acne, psoriasis, eczema, seborrheic dermatitis, and hidradenitis suppurativa. Reference citations in identified works also were reviewed. Interventional (experimental studies or clinical trials), survey-based, and observational studies that investigated the effects of low-glycemic or ketogenic diets for the treatment of inflammatory skin conditions were included. Inclusion criteria were studies assessing acne, psoriasis, SD, AD, and HS. Exclusion criteria were studies published before 1965; those written in languages other than English; and those analyzing other diets, such as the Mediterranean or low-fat diets. The search yielded a total of 11 observational studies and 4 controlled studies published between 1966 and January 2023. Because this analysis utilized publicly available data and did not qualify as human subject research, institutional review board approval was not required.

Results

Acne Vulgaris—Acne vulgaris is a disease of chronic pilosebaceous inflammation and follicular epithelial proliferation associated with Propionibacterium acnes. The association between acne and low-glycemic diets has been examined in several studies. Diet quality is measured and assessed using the glycemic index (GI), which is the effect of a single food on postprandial blood glucose, and the glycemic load, which is the GI adjusted for carbohydrates per serving.14 High levels of GI and glycemic load are associated with hyperinsulinemia and an increase in insulinlike growth factor 1 concentration that promotes mechanistic target of rapamycin (mTOR) complex 1–mediated follicular lipogenesis, sebum fatty acid production, and androgen synthesis.15Propionibacterium acnes directly activates toll-like receptor 2 on monocytes through damage-associated molecular patterns and indirectly through products of triglyceride catalysis, causing release of IL-12, IL-6, tumor necrosis factor α, and other proinflammatory cytokines.16 Therefore, lifestyle modifications focused on strict glucose control have been postulated to reduce acne severity via modulation of lipogenesis, androgen concentration, and inflammation.

Six survey-based studies evaluated sugar intake in patients with acne compared to healthy matched controls (eTable). Among these studies, 5 reported higher glycemic loads or daily sugar intake in acne patients compared to individuals without acne.12,19,20,26,28 The remaining study was conducted in 1967 and enrolled 16 acne patients and 32 matched controls. It reported no significant difference in sugar intake between the groups (P>.05).17

CT11302075_eTable_part1.jpg

CT11302075_eTable_part2.jpg

 

 

Smith et al18 randomized 43 male patients aged 15 to 25 years with facial acne into 2 cohorts for 12 weeks, each consuming either a low-glycemic diet (25% protein, 45% low-glycemic food [fruits, whole grains], and 30% fat) or a carbohydrate-dense diet of foods with medium to high GI based on prior documentation of the original diet. Patients were instructed to use a noncomedogenic cleanser as their only acne treatment. At 12 weeks, patients consuming the low-glycemic diet had an average of 23.5 fewer inflammatory lesions, while those in the intervention group had 12.0 fewer lesions (P=.03).18

In another controlled study by Kwon et al,21 32 male and female acne patients were randomized to a low-glycemic diet (25% protein, 45% low-glycemic food, and 30% fat) or a standard diet for 10 weeks. Patients on the low-glycemic diet experienced a 70.9% reduction in inflammatory lesions (P<.05). Hematoxylin and eosin staining and image analysis were performed to measure sebaceous gland surface area in the low-glycemic diet group, which decreased from 0.32 to 0.24 mm2 (P=.03). The sebaceous gland surface area in the control group was not reported. Moreover, patients on the low-glycemic diet had reduced IL-8 immunohistochemical staining (decreasing from 2.9 to 1.7 [P=.03]) and sterol regulatory element-binding protein 1 levels (decreasing from 2.6 to 1.3 [P=.03]), suggesting suppression of ongoing inflammation. Patients on the low-glycemic diet had no significant difference in transforming growth factor β1(P=.83). In the control group, there was no difference in IL-8, sterol regulatory element binding protein 1, or transforming growth factor β1 (P>.05) on immunohistochemical staining.21

Psoriasis—Psoriasis is a systemic inflammatory disease characterized by hyperproliferation and aberrant keratinocyte plaque formation. The innate immune response of keratinocytes in response to epidermal damage or infection begins with neutrophil recruitment and dendritic cell activation. Dendritic cell secretion of IL-23 promotes T-cell differentiation into helper T cells (TH1) that subsequently secrete IL-17 and IL-22, thereby stimulating keratinocyte proliferation and eventual plaque formation. The relationship between diet and psoriasis is poorly understood; however, hyperinsulinemia is associated with greater severity of psoriasis.31 

Four observational studies examined sugar intake in psoriasis patients. Barrea et al23 conducted a survey-based study of 82 male participants (41 with psoriasis and 41 healthy controls), reporting that PASI score was correlated with intake of simple carbohydrates (percentage of total kilocalorie)(r=0.564, P<.001). Another study by Yamashita et al27 found higher sugar intake in psoriasis patients than controls (P=.003) based on surveys from 70 patients with psoriasis and 70 matched healthy controls.

These findings contrast with 2 survey-based studies by Johnson et al22 and Afifi et al25 of sugar intake in psoriasis patients using the National Health and Nutrition Examination Survey. Johnson et al22 reported reduced sugar intake among 156 psoriasis patients compared with 6104 unmatched controls (odds ratio, 0.998; CI, 0.996-1 [P=.04]) from 2003 to 2006. Similarly, Afifi et al25 reported decreased sugar intake in 1206 psoriasis patients compared with sex- and age-matched controls (P<.0001) in 2009 and 2010. When patients were asked about dietary triggers, 13.8% of psoriasis patients reported sugar as the most common trigger, which was more frequent than alcohol (13.6%), gluten (7.2%), and dairy (6%).25

Castaldo et al29,30 published 2 nonrandomized clinical intervention studies in 2020 and 2021 evaluating the impact of the ketogenic diet on psoriasis. In the first study, 37 psoriasis patients followed a 10-week diet consisting of 4 weeks on a ketogenic diet (500 kcal/d) followed by 6 weeks on a low-caloric Mediterranean diet.29 At the end of the intervention, there was a 17.4% reduction in PASI score, a 33.2-point reduction in itch severity score, and a 13.4-point reduction in the dermatology life quality index score; however, this study did not include a control diet group for comparison.29 The second study included 30 psoriasis patients on a ketogenic diet and 30 control patients without psoriasis on a regular diet.30 The ketogenic diet consisted of 400 to 500 g of vegetables, 20 to 30 g of fat, and a proportion of protein based on body weight with at least 12 g of whey protein and various amino acids. Patients on the ketogenic diet had significant reduction in PASI scores (value relative to clinical features, 1.4916 [P=.007]). Furthermore, concentrations of cytokines IL-2 (P=.04) and IL-1β (P=.006) decreased following the ketogenic diet but were not measured in the control group.30

Seborrheic Dermatitis—Seborrheic dermatitis is associated with overcolonization of Malassezia species near lipid-rich sebaceous glands. Malassezia hydrolyzes free fatty acids, yielding oleic acids and leading to T-cell release of IL-8 and IL-17.32 Literature is sparse regarding how dietary modifications may play a role in disease severity. In a survey study, Bett et al17 compared 16 SD patients to 1:2 matched controls (N=29) to investigate the relationship between sugar consumption and presence of disease. Two control cohorts were selected, 1 from clinic patients diagnosed with verruca and 1 matched by age and sex from a survey-based study at a facility in London, England. Sugar intake was measured both in total grams per day and in “beverage sugar” per day, defined as sugar taken in tea and coffee. There was higher total sugar and higher beverage sugar intake among the SD group compared with both control groups (P<.05).17

 

 

Atopic Dermatitis—Atopic dermatitis is a disease of epidermal barrier dysfunction and IgE-mediated allergic sensitization.33 There are several mechanisms by which skin structure may be disrupted. It is well established that filaggrin mutations inhibit stratum corneum maturation and lamellar matrix deposition.34 Upregulation of IL-4–, IL-13–, and IL-17–secreting TH2 cells also is associated with disruption of tight junctions and reduction of filaggrin.35,36 Given that a T cell–mediated inflammatory response is involved in disease pathogenesis, glycemic control is hypothesized to have therapeutic potential.

Nosrati et al24 surveyed 169 AD patients about their perceived dietary triggers through a 61-question survey based on the National Health and Nutrition Examination Survey. Respondents were queried about their perceptions and dietary changes, such as removal or addition of specific food groups and trial of specific diets. Overall, 16.5% of patients reported sugar being a trigger, making it the fourth most common among those surveyed and less common than dairy (24.8%), gluten (18.3%), and alcohol (17.1%).24

Hidradenitis Suppurativa—Hidradenitis suppurativa is driven by hyperkeratosis, dilatation, and occlusion of pilosebaceous follicular ducts, whose eventual rupture evokes a local acute inflammatory response.37 The inciting event for both acne and HS involves mTOR complex–mediated follicular hyperproliferation andinsulinlike growth factor 1 stimulation of androgen receptors in pilosebaceous glands. Given the similarities between the pathogenesis of acne and HS, it is hypothesized that lifestyle changes, including diet modification, may have a beneficial effect on HS.38-40

Comment

Acne—Overall, there is strong evidence supporting the efficacy of a low-glycemic diet in the treatment of acne. Notably, among the 6 observational studies identified, there was 1 conflicting study by Bett et al17 that did not find a statistically significant difference in glucose intake between acne and control patients. However, this study included only 16 acne patients, whereas the other 5 observational studies included 32 to 2255 patients.17 The strongest evidence supporting low-glycemic dietary interventions in acne treatment is from 2 rigorous randomized clinical trials by Kwon et al21 and Smith et al.18 These trials used intention-to-treat models and maintained consistency in gender, age, and acne treatment protocols across both control and treatment groups. To ensure compliance with dietary interventions, daily telephone calls, food logs, and 24-hour urea sampling were utilized. Acne outcomes were assessed by a dermatologist who remained blinded with well-defined outcome measures. An important limitation of these studies is the difficulty in attributing the observed results solely to reduced glucose intake, as low-glycemic diets often lead to other dietary changes, including reduced fat intake and increased nutrient consumption.18,21

A 2022 systematic review of acne by Meixiong et al41 further reinforced the beneficial effects of low-glycemic diets in the management of acne patients. The group reviewed 6 interventional studies and 28 observational studies to investigate the relationship among acne, dairy, and glycemic content and found an association between decreased glucose and dairy on reduction of acne.41

It is likely that the ketogenic diet, which limits glucose, would be beneficial for acne patients. There may be added benefit through elevated ketone bodies and substantially reduced insulin secretion. However, because there are no observational or interventional studies, further research is needed to draw firm conclusions regarding diet for acne treatment. A randomized clinical trial investigating the effects of the ketogenic diet compared to the low-glycemic diet compared to a regular diet would be valuable.

Psoriasis—Among psoriasis studies, there was a lack of consensus regarding glucose intake and correlation with disease. Among the 4 observational studies, 2 reported increased glucose intake among psoriasis patients and 2 reported decreased glucose intake. It is plausible that the variability in studies is due to differences in sample size and diet heterogeneity among study populations. More specifically, Johnson et al22 and Afifi et al25 analyzed large sample sizes of 6260 and 2412 US participants, respectively, and found decreased sugar intake among psoriasis patients compared to controls. In comparison, Barrea et al23 and Yamashita et al27 analyzed substantially smaller and more specific populations consisting of 82 Italian and 140 Japanese participants, respectively; both reported increased glucose intake among psoriasis patients compared to controls. These seemingly antithetical results may be explained by regional dietary differences, with varying proportions of meats, vegetables, antioxidants, and vitamins.

 

 

Moreover, the variation among studies may be further explained by the high prevalence of comorbidities among psoriasis patients. In the study by Barrea et al,23 psoriasis patients had higher fasting glucose (P=.004) and insulin (P=.022) levels than healthy patients. After adjusting for body mass index and metabolic syndrome, the correlation coefficient measuring the relationship between the PASI score and intake of simple carbohydrates changed from r=0.564 (P<.001) to r=0.352 (P=.028). The confounding impact of these comorbidities was further highlighted by Yamashita et al,27 who found statistically significant differences in glucose intake between psoriasis and healthy patients (P=.003). However, they reported diminished significance on additional subgroup analysis accounting for potential comorbidities (P=.994).27 Johnson et al22 and Afifi et al25 did not account for comorbidities; therefore, the 4 observational study results must be interpreted cautiously.

The 2 randomized clinical trials by Castaldo et al29,30 weakly suggest that a ketogenic diet may be beneficial for psoriasis patients. The studies have several notable limitations, including insufficient sample sizes and control groups. Thus, the decreased PASI scores reported in psoriasis patients on the ketogenic diets are challenging to interpret. Additionally, both studies placed patients on highly restrictive diets of 500 kcal/d for 4 weeks. The feasibility of recommending such a diet to patients in clinical practice is questionable. Diets of less than 500 kcal/d may be dangerous for patients with underlying comorbidities and are unlikely to serve as long-term solutions.23 To contextualize our findings, a 2022 review by Chung et al42 examined the impact of various diets—low-caloric, gluten-free, Mediterranean, Western, and ketogenic—on psoriasis and reported insufficient evidence to suggest a benefit to the ketogenic diet for psoriasis patients, though the Mediterranean diet may be well suited for psoriasis patients because of improved cardiovascular health and reduced mortality.

Seborrheic Dermatitis—Sanders et al43 found that patients with a high-fruit diet had lower odds of having SD, while those on a Western diet had higher odds of having SD. Although the study did not measure glycemic load, it is conceivable that the high glycemic load characteristic of the Western diet contributed to these findings.43 However, no studies have investigated the direct link between low-glycemic or ketogenic diets and SD, leaving this area open for further study.

Atopic Dermatitis—It has been hypothesized that mitigating T cell–mediated inflammation via glucose control may contribute to the improvement in AD.35,36 However, in one study, 16.5% of AD patients self-identified sugar as a dietary trigger, ranking fourth among other dietary triggers.24 Thus, the connection between glucose levels and AD warrants further exploration.

Hidradenitis Suppurativa—Given the role of metabolic and hormonal influence in HS as well as the overlapping pathophysiology with acne, it is possible that low-glycemic and ketogenic diets may have a role in improving HS.38-40 However, there is a gap in observation and controlled studies investigating the link between low-glycemic or ketogenic diets and HS.

Conclusion

Our analysis focused on interventional and observational research exploring the effects of low-glycemic and ketogenic diets on associations and treatment of inflammatory skin conditions. There is sufficient evidence to counsel acne patients on the benefits of a low-glycemic diet as an adjunctive treatment for acne. Currently, there is insufficient evidence to recommend a low-glycemic or ketogenic diet as a treatment for patients with any other inflammatory skin disease. Prospective and controlled clinical trials are needed to clarify the utility of dietary interventions for treating inflammatory skin conditions.

References
  1. Pickett K, Loveman E, Kalita N, et al. Educational interventions to improve quality of life in people with chronic inflammatory skin diseases: systematic reviews of clinical effectiveness and cost-effectiveness. Health Technol Assess. 2015;19:1-176, v-vi.
  2. Giugliano D, Ceriello A, Esposito K. The effects of diet on inflammation: emphasis on the metabolic syndrome. J Am Coll Cardiol. 2006;48:677-685.
  3. Dowlatshahi EA, van der Voort EA, Arends LR, et al. Markers of systemic inflammation in psoriasis: a systematic review and meta-analysis. Br J Dermatol. 2013;169:266-282.
  4. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263-269.
  5. Melnik BC. Acne vulgaris: the metabolic syndrome of the pilosebaceous follicle. Clin Dermatol. 2018;36:29-40.
  6. Upala S, Sanguankeo A. Effect of lifestyle weight loss intervention on disease severity in patients with psoriasis: a systematic review and meta-analysis. Int J Obes (Lond). 2015;39:1197-1202.
  7. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020;10:5754.
  8. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacol Physiol. 2012;25:111-117.
  9. Masood W, Annamaraju P, Khan Suheb MZ, et al. Ketogenic diet. StatPearls. StatPearls Publishing; 2023.
  10. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. J Dermatolog Treat. 2017;28:484-487.
  11. Zhang D, Jin W, Wu R, et al. High glucose intake exacerbates autoimmunity through reactive-oxygen-species-mediated TGF-β cytokine activation. Immunity. 2019;51:671-681.e5.
  12. Cerman AA, Aktas E, Altunay IK, et al. Dietary glycemic factors, insulin resistance, and adiponectin levels in acne vulgaris. J Am Acad Dermatol. 2016;75:155-162.
  13. Ferrere G, Tidjani Alou M, Liu P, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6:e145207.
  14. Burris J, Shikany JM, Rietkerk W, et al. A Low glycemic index and glycemic load diet decreases insulin-like growth factor-1 among adults with moderate and severe acne: a short-duration, 2-week randomized controlled trial. J Acad Nutr Diet. 2018;118:1874-1885.
  15. Tan JKL, Stein Gold LF, Alexis AF, et al. Current concepts in acne pathogenesis: pathways to inflammation. Semin Cutan Med Surg. 2018;37(3S):S60-S62.
  16. Kim J, Ochoa MT, Krutzik SR, et al. Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses. J Immunol. 2002;169:1535-1541.
  17. Bett DG, Morland J, Yudkin J. Sugar consumption in acne vulgaris and seborrhoeic dermatitis. Br Med J. 1967;3:153-155.
  18. Smith RN, Mann NJ, Braue A, et al. A low-glycemic-load diet improves symptoms in acne vulgaris patients: a randomized controlled trial. Am J Clin Nutr. 2007;86:107-115.
  19. Rouhani P, Berman B, Rouhani G. Acne improves with a popular, low glycemic diet from South Beach. J Am Acad Dermatol. 2009;60(Suppl 1):AB14.
  20. Aksu AE, Metintas S, Saracoglu ZN, et al. Acne: prevalence and relationship with dietary habits in Eskisehir, Turkey. J Eur Acad Dermatol Venereol. 2012;26:1503-1509.
  21. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. Acta Derm Venereol. 2012;92:241-246.
  22. Johnson JA, Ma C, Kanada KN, et al. Diet and nutrition in psoriasis: analysis of the National Health and Nutrition Examination Survey (NHANES) in the United States. J Eur Acad Dermatol Venereol. 2014;28:327-332.
  23. Barrea L, Macchia PE, Tarantino G, et al. Nutrition: a key environmental dietary factor in clinical severity and cardio-metabolic risk in psoriatic male patients evaluated by 7-day food-frequency questionnaire. J Transl Med. 2015;13:303.
  24. Nosrati A, Afifi L, Danesh MJ, et al. Dietary modifications in atopic dermatitis: patient-reported outcomes. J Dermatolog Treat. 2017;28:523-538.
  25. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. Dermatol Ther (Heidelb). 2017;7:227-242.
  26. Burris J, Rietkerk W, Shikany JM, et al. Differences in dietary glycemic load and hormones in New York City adults with no and moderate/severe acne. J Acad Nutr Diet. 2017;117:1375-1383.
  27. Yamashita H, Morita T, Ito M, et al. Dietary habits in Japanese patients with psoriasis and psoriatic arthritis: low intake of meat in psoriasis and high intake of vitamin A in psoriatic arthritis. J Dermatol. 2019;46:759-769.
  28. Marson J, Baldwin HE. 12761 Acne, twins, and glycemic index: a sweet pilot study of diet and dietary beliefs. J Am Acad Dermatol. 2020;83(Suppl):AB110.
  29. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition. 2020;74:110757.
  30. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res. 2021;20:1509-1521.
  31. Ip W, Kirchhof MG. Glycemic control in the treatment of psoriasis. Dermatology. 2017;233:23-29.
  32. Vijaya Chandra SH, Srinivas R, Dawson TL Jr, et al. Cutaneous Malassezia: commensal, pathogen, or protector? Front Cell Infect Microbiol. 2020;10:614446.
  33. David Boothe W, Tarbox JA, Tarbox MB. Atopic dermatitis: pathophysiology. Adv Exp Med Biol. 2017;1027:21-37.
  34. Guttman-Yassky E, Hanifin JM, Boguniewicz M, et al. The role of phosphodiesterase 4 in the pathophysiology of atopic dermatitis and the perspective for its inhibition. Exp Dermatol. 2019;28:3-10.
  35. Furue K, Ito T, Tsuji G, et al. The IL-13–OVOL1–FLG axis in atopic dermatitis. Immunology. 2019;158:281-286.
  36. Renert-Yuval Y, Guttman-Yassky E. New treatments for atopic dermatitis targeting beyond IL-4/IL-13 cytokines. Ann Allergy Asthma Immunol. 2020;124:28-35.
  37. Sellheyer K, Krahl D. “Hidradenitis suppurativa” is acne inversa! An appeal to (finally) abandon a misnomer. Int J Dermatol. 2005;44:535-540.
  38. Danby FW, Margesson LJ. Hidradenitis suppurativa. Dermatol Clin. 2010;28:779-793.
  39. Fernandez JM, Marr KD, Hendricks AJ, et al. Alleviating and exacerbating foods in hidradenitis suppurativa. Dermatol Ther. 2020;33:E14246.
  40. Yamanaka-Takaichi M, Revankar R, Shih T, et al. Expert consensus on priority research gaps in dietary and lifestyle factors in hidradenitis suppurativa: a Delphi consensus study. Arch Dermatol Res. 2023;315:2129-2136.
  41. Meixiong J, Ricco C, Vasavda C, et al. Diet and acne: a systematic review. JAAD Int. 2022;7:95-112.
  42. Chung M, Bartholomew E, Yeroushalmi S, et al. Dietary intervention and supplements in the management of psoriasis: current perspectives. Psoriasis (Auckland). 2022;12:151-176. doi:10.2147/PTT.S328581
  43. Sanders MGH, Pardo LM, Ginger RS, et al. Association between diet and seborrheic dermatitis: a cross-sectional study. J Invest Dermatol. 2019;139:108-114.
Article PDF
Author and Disclosure Information

Katie Roster, Lillian Xie, and Terry Nguyen are from New York Medical College, Valhalla. Dr. Lipner is from the Department of Dermatology,Weill Cornell Medicine, New York, New York.

Katie Roster, Lillian Xie, and Terry Nguyen report no conflict of interest. Dr. Lipner has been a consultant for Ortho Dermatologics; has received research grants from BelleTorus Corporation and Moberg Pharma; and has served on the board for Hoth Therapeutics.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).

Issue
Cutis - 113(2)
Publications
Topics
Page Number
75-80,E1-E2
Sections
Author and Disclosure Information

Katie Roster, Lillian Xie, and Terry Nguyen are from New York Medical College, Valhalla. Dr. Lipner is from the Department of Dermatology,Weill Cornell Medicine, New York, New York.

Katie Roster, Lillian Xie, and Terry Nguyen report no conflict of interest. Dr. Lipner has been a consultant for Ortho Dermatologics; has received research grants from BelleTorus Corporation and Moberg Pharma; and has served on the board for Hoth Therapeutics.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).

Author and Disclosure Information

Katie Roster, Lillian Xie, and Terry Nguyen are from New York Medical College, Valhalla. Dr. Lipner is from the Department of Dermatology,Weill Cornell Medicine, New York, New York.

Katie Roster, Lillian Xie, and Terry Nguyen report no conflict of interest. Dr. Lipner has been a consultant for Ortho Dermatologics; has received research grants from BelleTorus Corporation and Moberg Pharma; and has served on the board for Hoth Therapeutics.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).

Article PDF
Article PDF

Inflammatory skin conditions often have a relapsing and remitting course and represent a large proportion of chronic skin diseases. Common inflammatory skin disorders include acne, psoriasis, hidradenitis suppurativa (HS), atopic dermatitis (AD), and seborrheic dermatitis (SD).1 Although each of these conditions has a unique pathogenesis, they all are driven by a background of chronic inflammation. It has been reported that diets with high levels of refined carbohydrates and saturated or trans-fatty acids may exacerbate existing inflammation.2 Consequently, dietary interventions, such as the ketogenic and low-glycemic diets, have potential anti-inflammatory and metabolic effects that are being assessed as stand-alone or adjunctive therapies for dermatologic diseases.

Diet may partially influence systemic inflammation through its effect on weight. Higher body mass index and obesity are linked to a low-grade inflammatory state and higher levels of circulating inflammatory markers. Therefore, weight loss leads to decreases in inflammatory cytokines, including C-reactive protein, tumor necrosis factor α, and IL-6.3 These cytokines and metabolic effects overlap with inflammatory skin condition pathways. It also is posited that decreased insulin release associated with weight loss results in decreased sebaceous lipogenesis and androgens, which drive keratinocyte proliferation and acne development.4,5 For instance, in a 2015 meta-analysis of 5 randomized controlled trials on psoriasis, patients in the weight loss intervention group had more substantial reductions in psoriasis area and severity index (PASI) scores compared with controls receiving usual care (P=.004).6 However, in a systematic review of 35 studies on acne vulgaris, overweight and obese patients (defined by a body mass index of ≥23 kg/m2) had similar odds of having acne compared with normal-weight individuals (P=.671).7

Similar to weight loss, ketogenesis acts as a negative feedback mechanism to reduce insulin release, leading to decreased inflammation and androgens that often exacerbate inflammatory skin diseases.8 Ketogenesis ensues when daily carbohydrate intake is limited to less than 50 g, and long-term adherence to a ketogenic diet results in metabolic reliance on ketone bodies such as acetoacetate, β-hydroxybutyrate, and acetone.9 These metabolites may decrease free radical damage and consequently improve signs and symptoms of acne, psoriasis, and other inflammatory skin diseases.10-12 Similarly, increased ketones also may decrease activation of the NLRP3 (NOD-, LRR-, and Pyrin domain-containing protein 3) inflammasome and therefore reduce inflammatory markers such as IL-1β and IL-1.4,13 Several proposed mechanisms are outlined in the Table.

CT11302075_Table.jpg

Collectively, low-glycemic and ketogenic diets have been proposed as potential interventions for reducing inflammatory skin conditions. These dietary approaches are hypothesized to exert their effects by facilitating weight loss, elevating ketone levels, and reducing systemic inflammation. The current review summarizes the existing evidence on ketogenic and low-glycemic diets as treatments for inflammatory skin conditions and evaluates the potential benefits of these dietary interventions in managing and improving outcomes for individuals with inflammatory skin conditions.

Methods

Using PubMed for articles indexed for MEDLINE and Google Scholar, a review of the literature was conducted with a combination of the following search terms: low-glycemic diet, inflammatory, dermatologic, ketogenic diet, inflammation, dermatology, acne, psoriasis, eczema, seborrheic dermatitis, and hidradenitis suppurativa. Reference citations in identified works also were reviewed. Interventional (experimental studies or clinical trials), survey-based, and observational studies that investigated the effects of low-glycemic or ketogenic diets for the treatment of inflammatory skin conditions were included. Inclusion criteria were studies assessing acne, psoriasis, SD, AD, and HS. Exclusion criteria were studies published before 1965; those written in languages other than English; and those analyzing other diets, such as the Mediterranean or low-fat diets. The search yielded a total of 11 observational studies and 4 controlled studies published between 1966 and January 2023. Because this analysis utilized publicly available data and did not qualify as human subject research, institutional review board approval was not required.

Results

Acne Vulgaris—Acne vulgaris is a disease of chronic pilosebaceous inflammation and follicular epithelial proliferation associated with Propionibacterium acnes. The association between acne and low-glycemic diets has been examined in several studies. Diet quality is measured and assessed using the glycemic index (GI), which is the effect of a single food on postprandial blood glucose, and the glycemic load, which is the GI adjusted for carbohydrates per serving.14 High levels of GI and glycemic load are associated with hyperinsulinemia and an increase in insulinlike growth factor 1 concentration that promotes mechanistic target of rapamycin (mTOR) complex 1–mediated follicular lipogenesis, sebum fatty acid production, and androgen synthesis.15Propionibacterium acnes directly activates toll-like receptor 2 on monocytes through damage-associated molecular patterns and indirectly through products of triglyceride catalysis, causing release of IL-12, IL-6, tumor necrosis factor α, and other proinflammatory cytokines.16 Therefore, lifestyle modifications focused on strict glucose control have been postulated to reduce acne severity via modulation of lipogenesis, androgen concentration, and inflammation.

Six survey-based studies evaluated sugar intake in patients with acne compared to healthy matched controls (eTable). Among these studies, 5 reported higher glycemic loads or daily sugar intake in acne patients compared to individuals without acne.12,19,20,26,28 The remaining study was conducted in 1967 and enrolled 16 acne patients and 32 matched controls. It reported no significant difference in sugar intake between the groups (P>.05).17

CT11302075_eTable_part1.jpg

CT11302075_eTable_part2.jpg

 

 

Smith et al18 randomized 43 male patients aged 15 to 25 years with facial acne into 2 cohorts for 12 weeks, each consuming either a low-glycemic diet (25% protein, 45% low-glycemic food [fruits, whole grains], and 30% fat) or a carbohydrate-dense diet of foods with medium to high GI based on prior documentation of the original diet. Patients were instructed to use a noncomedogenic cleanser as their only acne treatment. At 12 weeks, patients consuming the low-glycemic diet had an average of 23.5 fewer inflammatory lesions, while those in the intervention group had 12.0 fewer lesions (P=.03).18

In another controlled study by Kwon et al,21 32 male and female acne patients were randomized to a low-glycemic diet (25% protein, 45% low-glycemic food, and 30% fat) or a standard diet for 10 weeks. Patients on the low-glycemic diet experienced a 70.9% reduction in inflammatory lesions (P<.05). Hematoxylin and eosin staining and image analysis were performed to measure sebaceous gland surface area in the low-glycemic diet group, which decreased from 0.32 to 0.24 mm2 (P=.03). The sebaceous gland surface area in the control group was not reported. Moreover, patients on the low-glycemic diet had reduced IL-8 immunohistochemical staining (decreasing from 2.9 to 1.7 [P=.03]) and sterol regulatory element-binding protein 1 levels (decreasing from 2.6 to 1.3 [P=.03]), suggesting suppression of ongoing inflammation. Patients on the low-glycemic diet had no significant difference in transforming growth factor β1(P=.83). In the control group, there was no difference in IL-8, sterol regulatory element binding protein 1, or transforming growth factor β1 (P>.05) on immunohistochemical staining.21

Psoriasis—Psoriasis is a systemic inflammatory disease characterized by hyperproliferation and aberrant keratinocyte plaque formation. The innate immune response of keratinocytes in response to epidermal damage or infection begins with neutrophil recruitment and dendritic cell activation. Dendritic cell secretion of IL-23 promotes T-cell differentiation into helper T cells (TH1) that subsequently secrete IL-17 and IL-22, thereby stimulating keratinocyte proliferation and eventual plaque formation. The relationship between diet and psoriasis is poorly understood; however, hyperinsulinemia is associated with greater severity of psoriasis.31 

Four observational studies examined sugar intake in psoriasis patients. Barrea et al23 conducted a survey-based study of 82 male participants (41 with psoriasis and 41 healthy controls), reporting that PASI score was correlated with intake of simple carbohydrates (percentage of total kilocalorie)(r=0.564, P<.001). Another study by Yamashita et al27 found higher sugar intake in psoriasis patients than controls (P=.003) based on surveys from 70 patients with psoriasis and 70 matched healthy controls.

These findings contrast with 2 survey-based studies by Johnson et al22 and Afifi et al25 of sugar intake in psoriasis patients using the National Health and Nutrition Examination Survey. Johnson et al22 reported reduced sugar intake among 156 psoriasis patients compared with 6104 unmatched controls (odds ratio, 0.998; CI, 0.996-1 [P=.04]) from 2003 to 2006. Similarly, Afifi et al25 reported decreased sugar intake in 1206 psoriasis patients compared with sex- and age-matched controls (P<.0001) in 2009 and 2010. When patients were asked about dietary triggers, 13.8% of psoriasis patients reported sugar as the most common trigger, which was more frequent than alcohol (13.6%), gluten (7.2%), and dairy (6%).25

Castaldo et al29,30 published 2 nonrandomized clinical intervention studies in 2020 and 2021 evaluating the impact of the ketogenic diet on psoriasis. In the first study, 37 psoriasis patients followed a 10-week diet consisting of 4 weeks on a ketogenic diet (500 kcal/d) followed by 6 weeks on a low-caloric Mediterranean diet.29 At the end of the intervention, there was a 17.4% reduction in PASI score, a 33.2-point reduction in itch severity score, and a 13.4-point reduction in the dermatology life quality index score; however, this study did not include a control diet group for comparison.29 The second study included 30 psoriasis patients on a ketogenic diet and 30 control patients without psoriasis on a regular diet.30 The ketogenic diet consisted of 400 to 500 g of vegetables, 20 to 30 g of fat, and a proportion of protein based on body weight with at least 12 g of whey protein and various amino acids. Patients on the ketogenic diet had significant reduction in PASI scores (value relative to clinical features, 1.4916 [P=.007]). Furthermore, concentrations of cytokines IL-2 (P=.04) and IL-1β (P=.006) decreased following the ketogenic diet but were not measured in the control group.30

Seborrheic Dermatitis—Seborrheic dermatitis is associated with overcolonization of Malassezia species near lipid-rich sebaceous glands. Malassezia hydrolyzes free fatty acids, yielding oleic acids and leading to T-cell release of IL-8 and IL-17.32 Literature is sparse regarding how dietary modifications may play a role in disease severity. In a survey study, Bett et al17 compared 16 SD patients to 1:2 matched controls (N=29) to investigate the relationship between sugar consumption and presence of disease. Two control cohorts were selected, 1 from clinic patients diagnosed with verruca and 1 matched by age and sex from a survey-based study at a facility in London, England. Sugar intake was measured both in total grams per day and in “beverage sugar” per day, defined as sugar taken in tea and coffee. There was higher total sugar and higher beverage sugar intake among the SD group compared with both control groups (P<.05).17

 

 

Atopic Dermatitis—Atopic dermatitis is a disease of epidermal barrier dysfunction and IgE-mediated allergic sensitization.33 There are several mechanisms by which skin structure may be disrupted. It is well established that filaggrin mutations inhibit stratum corneum maturation and lamellar matrix deposition.34 Upregulation of IL-4–, IL-13–, and IL-17–secreting TH2 cells also is associated with disruption of tight junctions and reduction of filaggrin.35,36 Given that a T cell–mediated inflammatory response is involved in disease pathogenesis, glycemic control is hypothesized to have therapeutic potential.

Nosrati et al24 surveyed 169 AD patients about their perceived dietary triggers through a 61-question survey based on the National Health and Nutrition Examination Survey. Respondents were queried about their perceptions and dietary changes, such as removal or addition of specific food groups and trial of specific diets. Overall, 16.5% of patients reported sugar being a trigger, making it the fourth most common among those surveyed and less common than dairy (24.8%), gluten (18.3%), and alcohol (17.1%).24

Hidradenitis Suppurativa—Hidradenitis suppurativa is driven by hyperkeratosis, dilatation, and occlusion of pilosebaceous follicular ducts, whose eventual rupture evokes a local acute inflammatory response.37 The inciting event for both acne and HS involves mTOR complex–mediated follicular hyperproliferation andinsulinlike growth factor 1 stimulation of androgen receptors in pilosebaceous glands. Given the similarities between the pathogenesis of acne and HS, it is hypothesized that lifestyle changes, including diet modification, may have a beneficial effect on HS.38-40

Comment

Acne—Overall, there is strong evidence supporting the efficacy of a low-glycemic diet in the treatment of acne. Notably, among the 6 observational studies identified, there was 1 conflicting study by Bett et al17 that did not find a statistically significant difference in glucose intake between acne and control patients. However, this study included only 16 acne patients, whereas the other 5 observational studies included 32 to 2255 patients.17 The strongest evidence supporting low-glycemic dietary interventions in acne treatment is from 2 rigorous randomized clinical trials by Kwon et al21 and Smith et al.18 These trials used intention-to-treat models and maintained consistency in gender, age, and acne treatment protocols across both control and treatment groups. To ensure compliance with dietary interventions, daily telephone calls, food logs, and 24-hour urea sampling were utilized. Acne outcomes were assessed by a dermatologist who remained blinded with well-defined outcome measures. An important limitation of these studies is the difficulty in attributing the observed results solely to reduced glucose intake, as low-glycemic diets often lead to other dietary changes, including reduced fat intake and increased nutrient consumption.18,21

A 2022 systematic review of acne by Meixiong et al41 further reinforced the beneficial effects of low-glycemic diets in the management of acne patients. The group reviewed 6 interventional studies and 28 observational studies to investigate the relationship among acne, dairy, and glycemic content and found an association between decreased glucose and dairy on reduction of acne.41

It is likely that the ketogenic diet, which limits glucose, would be beneficial for acne patients. There may be added benefit through elevated ketone bodies and substantially reduced insulin secretion. However, because there are no observational or interventional studies, further research is needed to draw firm conclusions regarding diet for acne treatment. A randomized clinical trial investigating the effects of the ketogenic diet compared to the low-glycemic diet compared to a regular diet would be valuable.

Psoriasis—Among psoriasis studies, there was a lack of consensus regarding glucose intake and correlation with disease. Among the 4 observational studies, 2 reported increased glucose intake among psoriasis patients and 2 reported decreased glucose intake. It is plausible that the variability in studies is due to differences in sample size and diet heterogeneity among study populations. More specifically, Johnson et al22 and Afifi et al25 analyzed large sample sizes of 6260 and 2412 US participants, respectively, and found decreased sugar intake among psoriasis patients compared to controls. In comparison, Barrea et al23 and Yamashita et al27 analyzed substantially smaller and more specific populations consisting of 82 Italian and 140 Japanese participants, respectively; both reported increased glucose intake among psoriasis patients compared to controls. These seemingly antithetical results may be explained by regional dietary differences, with varying proportions of meats, vegetables, antioxidants, and vitamins.

 

 

Moreover, the variation among studies may be further explained by the high prevalence of comorbidities among psoriasis patients. In the study by Barrea et al,23 psoriasis patients had higher fasting glucose (P=.004) and insulin (P=.022) levels than healthy patients. After adjusting for body mass index and metabolic syndrome, the correlation coefficient measuring the relationship between the PASI score and intake of simple carbohydrates changed from r=0.564 (P<.001) to r=0.352 (P=.028). The confounding impact of these comorbidities was further highlighted by Yamashita et al,27 who found statistically significant differences in glucose intake between psoriasis and healthy patients (P=.003). However, they reported diminished significance on additional subgroup analysis accounting for potential comorbidities (P=.994).27 Johnson et al22 and Afifi et al25 did not account for comorbidities; therefore, the 4 observational study results must be interpreted cautiously.

The 2 randomized clinical trials by Castaldo et al29,30 weakly suggest that a ketogenic diet may be beneficial for psoriasis patients. The studies have several notable limitations, including insufficient sample sizes and control groups. Thus, the decreased PASI scores reported in psoriasis patients on the ketogenic diets are challenging to interpret. Additionally, both studies placed patients on highly restrictive diets of 500 kcal/d for 4 weeks. The feasibility of recommending such a diet to patients in clinical practice is questionable. Diets of less than 500 kcal/d may be dangerous for patients with underlying comorbidities and are unlikely to serve as long-term solutions.23 To contextualize our findings, a 2022 review by Chung et al42 examined the impact of various diets—low-caloric, gluten-free, Mediterranean, Western, and ketogenic—on psoriasis and reported insufficient evidence to suggest a benefit to the ketogenic diet for psoriasis patients, though the Mediterranean diet may be well suited for psoriasis patients because of improved cardiovascular health and reduced mortality.

Seborrheic Dermatitis—Sanders et al43 found that patients with a high-fruit diet had lower odds of having SD, while those on a Western diet had higher odds of having SD. Although the study did not measure glycemic load, it is conceivable that the high glycemic load characteristic of the Western diet contributed to these findings.43 However, no studies have investigated the direct link between low-glycemic or ketogenic diets and SD, leaving this area open for further study.

Atopic Dermatitis—It has been hypothesized that mitigating T cell–mediated inflammation via glucose control may contribute to the improvement in AD.35,36 However, in one study, 16.5% of AD patients self-identified sugar as a dietary trigger, ranking fourth among other dietary triggers.24 Thus, the connection between glucose levels and AD warrants further exploration.

Hidradenitis Suppurativa—Given the role of metabolic and hormonal influence in HS as well as the overlapping pathophysiology with acne, it is possible that low-glycemic and ketogenic diets may have a role in improving HS.38-40 However, there is a gap in observation and controlled studies investigating the link between low-glycemic or ketogenic diets and HS.

Conclusion

Our analysis focused on interventional and observational research exploring the effects of low-glycemic and ketogenic diets on associations and treatment of inflammatory skin conditions. There is sufficient evidence to counsel acne patients on the benefits of a low-glycemic diet as an adjunctive treatment for acne. Currently, there is insufficient evidence to recommend a low-glycemic or ketogenic diet as a treatment for patients with any other inflammatory skin disease. Prospective and controlled clinical trials are needed to clarify the utility of dietary interventions for treating inflammatory skin conditions.

Inflammatory skin conditions often have a relapsing and remitting course and represent a large proportion of chronic skin diseases. Common inflammatory skin disorders include acne, psoriasis, hidradenitis suppurativa (HS), atopic dermatitis (AD), and seborrheic dermatitis (SD).1 Although each of these conditions has a unique pathogenesis, they all are driven by a background of chronic inflammation. It has been reported that diets with high levels of refined carbohydrates and saturated or trans-fatty acids may exacerbate existing inflammation.2 Consequently, dietary interventions, such as the ketogenic and low-glycemic diets, have potential anti-inflammatory and metabolic effects that are being assessed as stand-alone or adjunctive therapies for dermatologic diseases.

Diet may partially influence systemic inflammation through its effect on weight. Higher body mass index and obesity are linked to a low-grade inflammatory state and higher levels of circulating inflammatory markers. Therefore, weight loss leads to decreases in inflammatory cytokines, including C-reactive protein, tumor necrosis factor α, and IL-6.3 These cytokines and metabolic effects overlap with inflammatory skin condition pathways. It also is posited that decreased insulin release associated with weight loss results in decreased sebaceous lipogenesis and androgens, which drive keratinocyte proliferation and acne development.4,5 For instance, in a 2015 meta-analysis of 5 randomized controlled trials on psoriasis, patients in the weight loss intervention group had more substantial reductions in psoriasis area and severity index (PASI) scores compared with controls receiving usual care (P=.004).6 However, in a systematic review of 35 studies on acne vulgaris, overweight and obese patients (defined by a body mass index of ≥23 kg/m2) had similar odds of having acne compared with normal-weight individuals (P=.671).7

Similar to weight loss, ketogenesis acts as a negative feedback mechanism to reduce insulin release, leading to decreased inflammation and androgens that often exacerbate inflammatory skin diseases.8 Ketogenesis ensues when daily carbohydrate intake is limited to less than 50 g, and long-term adherence to a ketogenic diet results in metabolic reliance on ketone bodies such as acetoacetate, β-hydroxybutyrate, and acetone.9 These metabolites may decrease free radical damage and consequently improve signs and symptoms of acne, psoriasis, and other inflammatory skin diseases.10-12 Similarly, increased ketones also may decrease activation of the NLRP3 (NOD-, LRR-, and Pyrin domain-containing protein 3) inflammasome and therefore reduce inflammatory markers such as IL-1β and IL-1.4,13 Several proposed mechanisms are outlined in the Table.

CT11302075_Table.jpg

Collectively, low-glycemic and ketogenic diets have been proposed as potential interventions for reducing inflammatory skin conditions. These dietary approaches are hypothesized to exert their effects by facilitating weight loss, elevating ketone levels, and reducing systemic inflammation. The current review summarizes the existing evidence on ketogenic and low-glycemic diets as treatments for inflammatory skin conditions and evaluates the potential benefits of these dietary interventions in managing and improving outcomes for individuals with inflammatory skin conditions.

Methods

Using PubMed for articles indexed for MEDLINE and Google Scholar, a review of the literature was conducted with a combination of the following search terms: low-glycemic diet, inflammatory, dermatologic, ketogenic diet, inflammation, dermatology, acne, psoriasis, eczema, seborrheic dermatitis, and hidradenitis suppurativa. Reference citations in identified works also were reviewed. Interventional (experimental studies or clinical trials), survey-based, and observational studies that investigated the effects of low-glycemic or ketogenic diets for the treatment of inflammatory skin conditions were included. Inclusion criteria were studies assessing acne, psoriasis, SD, AD, and HS. Exclusion criteria were studies published before 1965; those written in languages other than English; and those analyzing other diets, such as the Mediterranean or low-fat diets. The search yielded a total of 11 observational studies and 4 controlled studies published between 1966 and January 2023. Because this analysis utilized publicly available data and did not qualify as human subject research, institutional review board approval was not required.

Results

Acne Vulgaris—Acne vulgaris is a disease of chronic pilosebaceous inflammation and follicular epithelial proliferation associated with Propionibacterium acnes. The association between acne and low-glycemic diets has been examined in several studies. Diet quality is measured and assessed using the glycemic index (GI), which is the effect of a single food on postprandial blood glucose, and the glycemic load, which is the GI adjusted for carbohydrates per serving.14 High levels of GI and glycemic load are associated with hyperinsulinemia and an increase in insulinlike growth factor 1 concentration that promotes mechanistic target of rapamycin (mTOR) complex 1–mediated follicular lipogenesis, sebum fatty acid production, and androgen synthesis.15Propionibacterium acnes directly activates toll-like receptor 2 on monocytes through damage-associated molecular patterns and indirectly through products of triglyceride catalysis, causing release of IL-12, IL-6, tumor necrosis factor α, and other proinflammatory cytokines.16 Therefore, lifestyle modifications focused on strict glucose control have been postulated to reduce acne severity via modulation of lipogenesis, androgen concentration, and inflammation.

Six survey-based studies evaluated sugar intake in patients with acne compared to healthy matched controls (eTable). Among these studies, 5 reported higher glycemic loads or daily sugar intake in acne patients compared to individuals without acne.12,19,20,26,28 The remaining study was conducted in 1967 and enrolled 16 acne patients and 32 matched controls. It reported no significant difference in sugar intake between the groups (P>.05).17

CT11302075_eTable_part1.jpg

CT11302075_eTable_part2.jpg

 

 

Smith et al18 randomized 43 male patients aged 15 to 25 years with facial acne into 2 cohorts for 12 weeks, each consuming either a low-glycemic diet (25% protein, 45% low-glycemic food [fruits, whole grains], and 30% fat) or a carbohydrate-dense diet of foods with medium to high GI based on prior documentation of the original diet. Patients were instructed to use a noncomedogenic cleanser as their only acne treatment. At 12 weeks, patients consuming the low-glycemic diet had an average of 23.5 fewer inflammatory lesions, while those in the intervention group had 12.0 fewer lesions (P=.03).18

In another controlled study by Kwon et al,21 32 male and female acne patients were randomized to a low-glycemic diet (25% protein, 45% low-glycemic food, and 30% fat) or a standard diet for 10 weeks. Patients on the low-glycemic diet experienced a 70.9% reduction in inflammatory lesions (P<.05). Hematoxylin and eosin staining and image analysis were performed to measure sebaceous gland surface area in the low-glycemic diet group, which decreased from 0.32 to 0.24 mm2 (P=.03). The sebaceous gland surface area in the control group was not reported. Moreover, patients on the low-glycemic diet had reduced IL-8 immunohistochemical staining (decreasing from 2.9 to 1.7 [P=.03]) and sterol regulatory element-binding protein 1 levels (decreasing from 2.6 to 1.3 [P=.03]), suggesting suppression of ongoing inflammation. Patients on the low-glycemic diet had no significant difference in transforming growth factor β1(P=.83). In the control group, there was no difference in IL-8, sterol regulatory element binding protein 1, or transforming growth factor β1 (P>.05) on immunohistochemical staining.21

Psoriasis—Psoriasis is a systemic inflammatory disease characterized by hyperproliferation and aberrant keratinocyte plaque formation. The innate immune response of keratinocytes in response to epidermal damage or infection begins with neutrophil recruitment and dendritic cell activation. Dendritic cell secretion of IL-23 promotes T-cell differentiation into helper T cells (TH1) that subsequently secrete IL-17 and IL-22, thereby stimulating keratinocyte proliferation and eventual plaque formation. The relationship between diet and psoriasis is poorly understood; however, hyperinsulinemia is associated with greater severity of psoriasis.31 

Four observational studies examined sugar intake in psoriasis patients. Barrea et al23 conducted a survey-based study of 82 male participants (41 with psoriasis and 41 healthy controls), reporting that PASI score was correlated with intake of simple carbohydrates (percentage of total kilocalorie)(r=0.564, P<.001). Another study by Yamashita et al27 found higher sugar intake in psoriasis patients than controls (P=.003) based on surveys from 70 patients with psoriasis and 70 matched healthy controls.

These findings contrast with 2 survey-based studies by Johnson et al22 and Afifi et al25 of sugar intake in psoriasis patients using the National Health and Nutrition Examination Survey. Johnson et al22 reported reduced sugar intake among 156 psoriasis patients compared with 6104 unmatched controls (odds ratio, 0.998; CI, 0.996-1 [P=.04]) from 2003 to 2006. Similarly, Afifi et al25 reported decreased sugar intake in 1206 psoriasis patients compared with sex- and age-matched controls (P<.0001) in 2009 and 2010. When patients were asked about dietary triggers, 13.8% of psoriasis patients reported sugar as the most common trigger, which was more frequent than alcohol (13.6%), gluten (7.2%), and dairy (6%).25

Castaldo et al29,30 published 2 nonrandomized clinical intervention studies in 2020 and 2021 evaluating the impact of the ketogenic diet on psoriasis. In the first study, 37 psoriasis patients followed a 10-week diet consisting of 4 weeks on a ketogenic diet (500 kcal/d) followed by 6 weeks on a low-caloric Mediterranean diet.29 At the end of the intervention, there was a 17.4% reduction in PASI score, a 33.2-point reduction in itch severity score, and a 13.4-point reduction in the dermatology life quality index score; however, this study did not include a control diet group for comparison.29 The second study included 30 psoriasis patients on a ketogenic diet and 30 control patients without psoriasis on a regular diet.30 The ketogenic diet consisted of 400 to 500 g of vegetables, 20 to 30 g of fat, and a proportion of protein based on body weight with at least 12 g of whey protein and various amino acids. Patients on the ketogenic diet had significant reduction in PASI scores (value relative to clinical features, 1.4916 [P=.007]). Furthermore, concentrations of cytokines IL-2 (P=.04) and IL-1β (P=.006) decreased following the ketogenic diet but were not measured in the control group.30

Seborrheic Dermatitis—Seborrheic dermatitis is associated with overcolonization of Malassezia species near lipid-rich sebaceous glands. Malassezia hydrolyzes free fatty acids, yielding oleic acids and leading to T-cell release of IL-8 and IL-17.32 Literature is sparse regarding how dietary modifications may play a role in disease severity. In a survey study, Bett et al17 compared 16 SD patients to 1:2 matched controls (N=29) to investigate the relationship between sugar consumption and presence of disease. Two control cohorts were selected, 1 from clinic patients diagnosed with verruca and 1 matched by age and sex from a survey-based study at a facility in London, England. Sugar intake was measured both in total grams per day and in “beverage sugar” per day, defined as sugar taken in tea and coffee. There was higher total sugar and higher beverage sugar intake among the SD group compared with both control groups (P<.05).17

 

 

Atopic Dermatitis—Atopic dermatitis is a disease of epidermal barrier dysfunction and IgE-mediated allergic sensitization.33 There are several mechanisms by which skin structure may be disrupted. It is well established that filaggrin mutations inhibit stratum corneum maturation and lamellar matrix deposition.34 Upregulation of IL-4–, IL-13–, and IL-17–secreting TH2 cells also is associated with disruption of tight junctions and reduction of filaggrin.35,36 Given that a T cell–mediated inflammatory response is involved in disease pathogenesis, glycemic control is hypothesized to have therapeutic potential.

Nosrati et al24 surveyed 169 AD patients about their perceived dietary triggers through a 61-question survey based on the National Health and Nutrition Examination Survey. Respondents were queried about their perceptions and dietary changes, such as removal or addition of specific food groups and trial of specific diets. Overall, 16.5% of patients reported sugar being a trigger, making it the fourth most common among those surveyed and less common than dairy (24.8%), gluten (18.3%), and alcohol (17.1%).24

Hidradenitis Suppurativa—Hidradenitis suppurativa is driven by hyperkeratosis, dilatation, and occlusion of pilosebaceous follicular ducts, whose eventual rupture evokes a local acute inflammatory response.37 The inciting event for both acne and HS involves mTOR complex–mediated follicular hyperproliferation andinsulinlike growth factor 1 stimulation of androgen receptors in pilosebaceous glands. Given the similarities between the pathogenesis of acne and HS, it is hypothesized that lifestyle changes, including diet modification, may have a beneficial effect on HS.38-40

Comment

Acne—Overall, there is strong evidence supporting the efficacy of a low-glycemic diet in the treatment of acne. Notably, among the 6 observational studies identified, there was 1 conflicting study by Bett et al17 that did not find a statistically significant difference in glucose intake between acne and control patients. However, this study included only 16 acne patients, whereas the other 5 observational studies included 32 to 2255 patients.17 The strongest evidence supporting low-glycemic dietary interventions in acne treatment is from 2 rigorous randomized clinical trials by Kwon et al21 and Smith et al.18 These trials used intention-to-treat models and maintained consistency in gender, age, and acne treatment protocols across both control and treatment groups. To ensure compliance with dietary interventions, daily telephone calls, food logs, and 24-hour urea sampling were utilized. Acne outcomes were assessed by a dermatologist who remained blinded with well-defined outcome measures. An important limitation of these studies is the difficulty in attributing the observed results solely to reduced glucose intake, as low-glycemic diets often lead to other dietary changes, including reduced fat intake and increased nutrient consumption.18,21

A 2022 systematic review of acne by Meixiong et al41 further reinforced the beneficial effects of low-glycemic diets in the management of acne patients. The group reviewed 6 interventional studies and 28 observational studies to investigate the relationship among acne, dairy, and glycemic content and found an association between decreased glucose and dairy on reduction of acne.41

It is likely that the ketogenic diet, which limits glucose, would be beneficial for acne patients. There may be added benefit through elevated ketone bodies and substantially reduced insulin secretion. However, because there are no observational or interventional studies, further research is needed to draw firm conclusions regarding diet for acne treatment. A randomized clinical trial investigating the effects of the ketogenic diet compared to the low-glycemic diet compared to a regular diet would be valuable.

Psoriasis—Among psoriasis studies, there was a lack of consensus regarding glucose intake and correlation with disease. Among the 4 observational studies, 2 reported increased glucose intake among psoriasis patients and 2 reported decreased glucose intake. It is plausible that the variability in studies is due to differences in sample size and diet heterogeneity among study populations. More specifically, Johnson et al22 and Afifi et al25 analyzed large sample sizes of 6260 and 2412 US participants, respectively, and found decreased sugar intake among psoriasis patients compared to controls. In comparison, Barrea et al23 and Yamashita et al27 analyzed substantially smaller and more specific populations consisting of 82 Italian and 140 Japanese participants, respectively; both reported increased glucose intake among psoriasis patients compared to controls. These seemingly antithetical results may be explained by regional dietary differences, with varying proportions of meats, vegetables, antioxidants, and vitamins.

 

 

Moreover, the variation among studies may be further explained by the high prevalence of comorbidities among psoriasis patients. In the study by Barrea et al,23 psoriasis patients had higher fasting glucose (P=.004) and insulin (P=.022) levels than healthy patients. After adjusting for body mass index and metabolic syndrome, the correlation coefficient measuring the relationship between the PASI score and intake of simple carbohydrates changed from r=0.564 (P<.001) to r=0.352 (P=.028). The confounding impact of these comorbidities was further highlighted by Yamashita et al,27 who found statistically significant differences in glucose intake between psoriasis and healthy patients (P=.003). However, they reported diminished significance on additional subgroup analysis accounting for potential comorbidities (P=.994).27 Johnson et al22 and Afifi et al25 did not account for comorbidities; therefore, the 4 observational study results must be interpreted cautiously.

The 2 randomized clinical trials by Castaldo et al29,30 weakly suggest that a ketogenic diet may be beneficial for psoriasis patients. The studies have several notable limitations, including insufficient sample sizes and control groups. Thus, the decreased PASI scores reported in psoriasis patients on the ketogenic diets are challenging to interpret. Additionally, both studies placed patients on highly restrictive diets of 500 kcal/d for 4 weeks. The feasibility of recommending such a diet to patients in clinical practice is questionable. Diets of less than 500 kcal/d may be dangerous for patients with underlying comorbidities and are unlikely to serve as long-term solutions.23 To contextualize our findings, a 2022 review by Chung et al42 examined the impact of various diets—low-caloric, gluten-free, Mediterranean, Western, and ketogenic—on psoriasis and reported insufficient evidence to suggest a benefit to the ketogenic diet for psoriasis patients, though the Mediterranean diet may be well suited for psoriasis patients because of improved cardiovascular health and reduced mortality.

Seborrheic Dermatitis—Sanders et al43 found that patients with a high-fruit diet had lower odds of having SD, while those on a Western diet had higher odds of having SD. Although the study did not measure glycemic load, it is conceivable that the high glycemic load characteristic of the Western diet contributed to these findings.43 However, no studies have investigated the direct link between low-glycemic or ketogenic diets and SD, leaving this area open for further study.

Atopic Dermatitis—It has been hypothesized that mitigating T cell–mediated inflammation via glucose control may contribute to the improvement in AD.35,36 However, in one study, 16.5% of AD patients self-identified sugar as a dietary trigger, ranking fourth among other dietary triggers.24 Thus, the connection between glucose levels and AD warrants further exploration.

Hidradenitis Suppurativa—Given the role of metabolic and hormonal influence in HS as well as the overlapping pathophysiology with acne, it is possible that low-glycemic and ketogenic diets may have a role in improving HS.38-40 However, there is a gap in observation and controlled studies investigating the link between low-glycemic or ketogenic diets and HS.

Conclusion

Our analysis focused on interventional and observational research exploring the effects of low-glycemic and ketogenic diets on associations and treatment of inflammatory skin conditions. There is sufficient evidence to counsel acne patients on the benefits of a low-glycemic diet as an adjunctive treatment for acne. Currently, there is insufficient evidence to recommend a low-glycemic or ketogenic diet as a treatment for patients with any other inflammatory skin disease. Prospective and controlled clinical trials are needed to clarify the utility of dietary interventions for treating inflammatory skin conditions.

References
  1. Pickett K, Loveman E, Kalita N, et al. Educational interventions to improve quality of life in people with chronic inflammatory skin diseases: systematic reviews of clinical effectiveness and cost-effectiveness. Health Technol Assess. 2015;19:1-176, v-vi.
  2. Giugliano D, Ceriello A, Esposito K. The effects of diet on inflammation: emphasis on the metabolic syndrome. J Am Coll Cardiol. 2006;48:677-685.
  3. Dowlatshahi EA, van der Voort EA, Arends LR, et al. Markers of systemic inflammation in psoriasis: a systematic review and meta-analysis. Br J Dermatol. 2013;169:266-282.
  4. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263-269.
  5. Melnik BC. Acne vulgaris: the metabolic syndrome of the pilosebaceous follicle. Clin Dermatol. 2018;36:29-40.
  6. Upala S, Sanguankeo A. Effect of lifestyle weight loss intervention on disease severity in patients with psoriasis: a systematic review and meta-analysis. Int J Obes (Lond). 2015;39:1197-1202.
  7. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020;10:5754.
  8. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacol Physiol. 2012;25:111-117.
  9. Masood W, Annamaraju P, Khan Suheb MZ, et al. Ketogenic diet. StatPearls. StatPearls Publishing; 2023.
  10. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. J Dermatolog Treat. 2017;28:484-487.
  11. Zhang D, Jin W, Wu R, et al. High glucose intake exacerbates autoimmunity through reactive-oxygen-species-mediated TGF-β cytokine activation. Immunity. 2019;51:671-681.e5.
  12. Cerman AA, Aktas E, Altunay IK, et al. Dietary glycemic factors, insulin resistance, and adiponectin levels in acne vulgaris. J Am Acad Dermatol. 2016;75:155-162.
  13. Ferrere G, Tidjani Alou M, Liu P, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6:e145207.
  14. Burris J, Shikany JM, Rietkerk W, et al. A Low glycemic index and glycemic load diet decreases insulin-like growth factor-1 among adults with moderate and severe acne: a short-duration, 2-week randomized controlled trial. J Acad Nutr Diet. 2018;118:1874-1885.
  15. Tan JKL, Stein Gold LF, Alexis AF, et al. Current concepts in acne pathogenesis: pathways to inflammation. Semin Cutan Med Surg. 2018;37(3S):S60-S62.
  16. Kim J, Ochoa MT, Krutzik SR, et al. Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses. J Immunol. 2002;169:1535-1541.
  17. Bett DG, Morland J, Yudkin J. Sugar consumption in acne vulgaris and seborrhoeic dermatitis. Br Med J. 1967;3:153-155.
  18. Smith RN, Mann NJ, Braue A, et al. A low-glycemic-load diet improves symptoms in acne vulgaris patients: a randomized controlled trial. Am J Clin Nutr. 2007;86:107-115.
  19. Rouhani P, Berman B, Rouhani G. Acne improves with a popular, low glycemic diet from South Beach. J Am Acad Dermatol. 2009;60(Suppl 1):AB14.
  20. Aksu AE, Metintas S, Saracoglu ZN, et al. Acne: prevalence and relationship with dietary habits in Eskisehir, Turkey. J Eur Acad Dermatol Venereol. 2012;26:1503-1509.
  21. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. Acta Derm Venereol. 2012;92:241-246.
  22. Johnson JA, Ma C, Kanada KN, et al. Diet and nutrition in psoriasis: analysis of the National Health and Nutrition Examination Survey (NHANES) in the United States. J Eur Acad Dermatol Venereol. 2014;28:327-332.
  23. Barrea L, Macchia PE, Tarantino G, et al. Nutrition: a key environmental dietary factor in clinical severity and cardio-metabolic risk in psoriatic male patients evaluated by 7-day food-frequency questionnaire. J Transl Med. 2015;13:303.
  24. Nosrati A, Afifi L, Danesh MJ, et al. Dietary modifications in atopic dermatitis: patient-reported outcomes. J Dermatolog Treat. 2017;28:523-538.
  25. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. Dermatol Ther (Heidelb). 2017;7:227-242.
  26. Burris J, Rietkerk W, Shikany JM, et al. Differences in dietary glycemic load and hormones in New York City adults with no and moderate/severe acne. J Acad Nutr Diet. 2017;117:1375-1383.
  27. Yamashita H, Morita T, Ito M, et al. Dietary habits in Japanese patients with psoriasis and psoriatic arthritis: low intake of meat in psoriasis and high intake of vitamin A in psoriatic arthritis. J Dermatol. 2019;46:759-769.
  28. Marson J, Baldwin HE. 12761 Acne, twins, and glycemic index: a sweet pilot study of diet and dietary beliefs. J Am Acad Dermatol. 2020;83(Suppl):AB110.
  29. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition. 2020;74:110757.
  30. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res. 2021;20:1509-1521.
  31. Ip W, Kirchhof MG. Glycemic control in the treatment of psoriasis. Dermatology. 2017;233:23-29.
  32. Vijaya Chandra SH, Srinivas R, Dawson TL Jr, et al. Cutaneous Malassezia: commensal, pathogen, or protector? Front Cell Infect Microbiol. 2020;10:614446.
  33. David Boothe W, Tarbox JA, Tarbox MB. Atopic dermatitis: pathophysiology. Adv Exp Med Biol. 2017;1027:21-37.
  34. Guttman-Yassky E, Hanifin JM, Boguniewicz M, et al. The role of phosphodiesterase 4 in the pathophysiology of atopic dermatitis and the perspective for its inhibition. Exp Dermatol. 2019;28:3-10.
  35. Furue K, Ito T, Tsuji G, et al. The IL-13–OVOL1–FLG axis in atopic dermatitis. Immunology. 2019;158:281-286.
  36. Renert-Yuval Y, Guttman-Yassky E. New treatments for atopic dermatitis targeting beyond IL-4/IL-13 cytokines. Ann Allergy Asthma Immunol. 2020;124:28-35.
  37. Sellheyer K, Krahl D. “Hidradenitis suppurativa” is acne inversa! An appeal to (finally) abandon a misnomer. Int J Dermatol. 2005;44:535-540.
  38. Danby FW, Margesson LJ. Hidradenitis suppurativa. Dermatol Clin. 2010;28:779-793.
  39. Fernandez JM, Marr KD, Hendricks AJ, et al. Alleviating and exacerbating foods in hidradenitis suppurativa. Dermatol Ther. 2020;33:E14246.
  40. Yamanaka-Takaichi M, Revankar R, Shih T, et al. Expert consensus on priority research gaps in dietary and lifestyle factors in hidradenitis suppurativa: a Delphi consensus study. Arch Dermatol Res. 2023;315:2129-2136.
  41. Meixiong J, Ricco C, Vasavda C, et al. Diet and acne: a systematic review. JAAD Int. 2022;7:95-112.
  42. Chung M, Bartholomew E, Yeroushalmi S, et al. Dietary intervention and supplements in the management of psoriasis: current perspectives. Psoriasis (Auckland). 2022;12:151-176. doi:10.2147/PTT.S328581
  43. Sanders MGH, Pardo LM, Ginger RS, et al. Association between diet and seborrheic dermatitis: a cross-sectional study. J Invest Dermatol. 2019;139:108-114.
References
  1. Pickett K, Loveman E, Kalita N, et al. Educational interventions to improve quality of life in people with chronic inflammatory skin diseases: systematic reviews of clinical effectiveness and cost-effectiveness. Health Technol Assess. 2015;19:1-176, v-vi.
  2. Giugliano D, Ceriello A, Esposito K. The effects of diet on inflammation: emphasis on the metabolic syndrome. J Am Coll Cardiol. 2006;48:677-685.
  3. Dowlatshahi EA, van der Voort EA, Arends LR, et al. Markers of systemic inflammation in psoriasis: a systematic review and meta-analysis. Br J Dermatol. 2013;169:266-282.
  4. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263-269.
  5. Melnik BC. Acne vulgaris: the metabolic syndrome of the pilosebaceous follicle. Clin Dermatol. 2018;36:29-40.
  6. Upala S, Sanguankeo A. Effect of lifestyle weight loss intervention on disease severity in patients with psoriasis: a systematic review and meta-analysis. Int J Obes (Lond). 2015;39:1197-1202.
  7. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020;10:5754.
  8. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. Skin Pharmacol Physiol. 2012;25:111-117.
  9. Masood W, Annamaraju P, Khan Suheb MZ, et al. Ketogenic diet. StatPearls. StatPearls Publishing; 2023.
  10. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. J Dermatolog Treat. 2017;28:484-487.
  11. Zhang D, Jin W, Wu R, et al. High glucose intake exacerbates autoimmunity through reactive-oxygen-species-mediated TGF-β cytokine activation. Immunity. 2019;51:671-681.e5.
  12. Cerman AA, Aktas E, Altunay IK, et al. Dietary glycemic factors, insulin resistance, and adiponectin levels in acne vulgaris. J Am Acad Dermatol. 2016;75:155-162.
  13. Ferrere G, Tidjani Alou M, Liu P, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. JCI Insight. 2021;6:e145207.
  14. Burris J, Shikany JM, Rietkerk W, et al. A Low glycemic index and glycemic load diet decreases insulin-like growth factor-1 among adults with moderate and severe acne: a short-duration, 2-week randomized controlled trial. J Acad Nutr Diet. 2018;118:1874-1885.
  15. Tan JKL, Stein Gold LF, Alexis AF, et al. Current concepts in acne pathogenesis: pathways to inflammation. Semin Cutan Med Surg. 2018;37(3S):S60-S62.
  16. Kim J, Ochoa MT, Krutzik SR, et al. Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses. J Immunol. 2002;169:1535-1541.
  17. Bett DG, Morland J, Yudkin J. Sugar consumption in acne vulgaris and seborrhoeic dermatitis. Br Med J. 1967;3:153-155.
  18. Smith RN, Mann NJ, Braue A, et al. A low-glycemic-load diet improves symptoms in acne vulgaris patients: a randomized controlled trial. Am J Clin Nutr. 2007;86:107-115.
  19. Rouhani P, Berman B, Rouhani G. Acne improves with a popular, low glycemic diet from South Beach. J Am Acad Dermatol. 2009;60(Suppl 1):AB14.
  20. Aksu AE, Metintas S, Saracoglu ZN, et al. Acne: prevalence and relationship with dietary habits in Eskisehir, Turkey. J Eur Acad Dermatol Venereol. 2012;26:1503-1509.
  21. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. Acta Derm Venereol. 2012;92:241-246.
  22. Johnson JA, Ma C, Kanada KN, et al. Diet and nutrition in psoriasis: analysis of the National Health and Nutrition Examination Survey (NHANES) in the United States. J Eur Acad Dermatol Venereol. 2014;28:327-332.
  23. Barrea L, Macchia PE, Tarantino G, et al. Nutrition: a key environmental dietary factor in clinical severity and cardio-metabolic risk in psoriatic male patients evaluated by 7-day food-frequency questionnaire. J Transl Med. 2015;13:303.
  24. Nosrati A, Afifi L, Danesh MJ, et al. Dietary modifications in atopic dermatitis: patient-reported outcomes. J Dermatolog Treat. 2017;28:523-538.
  25. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. Dermatol Ther (Heidelb). 2017;7:227-242.
  26. Burris J, Rietkerk W, Shikany JM, et al. Differences in dietary glycemic load and hormones in New York City adults with no and moderate/severe acne. J Acad Nutr Diet. 2017;117:1375-1383.
  27. Yamashita H, Morita T, Ito M, et al. Dietary habits in Japanese patients with psoriasis and psoriatic arthritis: low intake of meat in psoriasis and high intake of vitamin A in psoriatic arthritis. J Dermatol. 2019;46:759-769.
  28. Marson J, Baldwin HE. 12761 Acne, twins, and glycemic index: a sweet pilot study of diet and dietary beliefs. J Am Acad Dermatol. 2020;83(Suppl):AB110.
  29. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. Nutrition. 2020;74:110757.
  30. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. J Proteome Res. 2021;20:1509-1521.
  31. Ip W, Kirchhof MG. Glycemic control in the treatment of psoriasis. Dermatology. 2017;233:23-29.
  32. Vijaya Chandra SH, Srinivas R, Dawson TL Jr, et al. Cutaneous Malassezia: commensal, pathogen, or protector? Front Cell Infect Microbiol. 2020;10:614446.
  33. David Boothe W, Tarbox JA, Tarbox MB. Atopic dermatitis: pathophysiology. Adv Exp Med Biol. 2017;1027:21-37.
  34. Guttman-Yassky E, Hanifin JM, Boguniewicz M, et al. The role of phosphodiesterase 4 in the pathophysiology of atopic dermatitis and the perspective for its inhibition. Exp Dermatol. 2019;28:3-10.
  35. Furue K, Ito T, Tsuji G, et al. The IL-13–OVOL1–FLG axis in atopic dermatitis. Immunology. 2019;158:281-286.
  36. Renert-Yuval Y, Guttman-Yassky E. New treatments for atopic dermatitis targeting beyond IL-4/IL-13 cytokines. Ann Allergy Asthma Immunol. 2020;124:28-35.
  37. Sellheyer K, Krahl D. “Hidradenitis suppurativa” is acne inversa! An appeal to (finally) abandon a misnomer. Int J Dermatol. 2005;44:535-540.
  38. Danby FW, Margesson LJ. Hidradenitis suppurativa. Dermatol Clin. 2010;28:779-793.
  39. Fernandez JM, Marr KD, Hendricks AJ, et al. Alleviating and exacerbating foods in hidradenitis suppurativa. Dermatol Ther. 2020;33:E14246.
  40. Yamanaka-Takaichi M, Revankar R, Shih T, et al. Expert consensus on priority research gaps in dietary and lifestyle factors in hidradenitis suppurativa: a Delphi consensus study. Arch Dermatol Res. 2023;315:2129-2136.
  41. Meixiong J, Ricco C, Vasavda C, et al. Diet and acne: a systematic review. JAAD Int. 2022;7:95-112.
  42. Chung M, Bartholomew E, Yeroushalmi S, et al. Dietary intervention and supplements in the management of psoriasis: current perspectives. Psoriasis (Auckland). 2022;12:151-176. doi:10.2147/PTT.S328581
  43. Sanders MGH, Pardo LM, Ginger RS, et al. Association between diet and seborrheic dermatitis: a cross-sectional study. J Invest Dermatol. 2019;139:108-114.
Issue
Cutis - 113(2)
Issue
Cutis - 113(2)
Page Number
75-80,E1-E2
Page Number
75-80,E1-E2
Publications
Publications
Topics
Article Type
Display Headline
Impact of Ketogenic and Low-Glycemic Diets on Inflammatory Skin Conditions
Display Headline
Impact of Ketogenic and Low-Glycemic Diets on Inflammatory Skin Conditions
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Roster</fileName> <TBEID>0C02F16E.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F16E</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Roster</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240130T131947</firstPublished> <LastPublished>20240130T131947</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240130T131946</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Katie Roster, MS; Lillian Xie, BS; Terry Nguyen, MS</byline> <bylineText>Katie Roster, MS; Lillian Xie, BS; Terry Nguyen, MS; Shari R. Lipner, MD, PhD </bylineText> <bylineFull>Katie Roster, MS; Lillian Xie, BS; Terry Nguyen, MS</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>75-80,E1-E2</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Inflammatory skin conditions often have a relapsing and remitting course and represent a large proportion of chronic skin diseases. Common inflammatory skin dis</metaDescription> <articlePDF>300103</articlePDF> <teaserImage/> <title>Impact of Ketogenic and Low-Glycemic Diets on Inflammatory Skin Conditions</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>February</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>2</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>psoriasis</keyword> <keyword> acne</keyword> <keyword> atopic dermatitis</keyword> <keyword> autoimmune diseases</keyword> <keyword> AD</keyword> <keyword> inflammatory skin condition</keyword> <keyword> ketogenic diet</keyword> <keyword> low-glycemic diet</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>February 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term>171</term> <term>189</term> <term canonical="true">281</term> <term>29134</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026b5.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Impact of Ketogenic and Low-Glycemic Diets on Inflammatory Skin Conditions</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Diet plays an emerging role in dermatologic therapy. The ketogenic and low-glycemic diets have potential anti-inflammatory and metabolic effects, making them attractive for treating inflammatory skin conditions. We provide an overview of the current evidence on the effects of ketogenic and low-glycemic diets on inflammatory skin conditions including acne, psoriasis, seborrheic dermatitis (SD), atopic dermatitis (AD), and hidradenitis suppurativa (HS). We conclude that low-glycemic diets show promise for treating acne, while the evidence for ketogenic diets in treating other inflammatory skin conditions is limited. Randomized clinical trials are needed to explore the efficacy of these diets as stand-alone or adjunctive treatments for inflammatory skin conditions.</p> <p> <em><em>Cutis.</em> 2024;113:75-80, E1-E2.</em> </p> <p>Inflammatory skin conditions often have a relapsing and remitting course and represent a large proportion of chronic skin diseases. Common inflammatory skin disorders include acne, psoriasis, hidradenitis suppurativa (HS), atopic dermatitis (AD), and seborrheic dermatitis (SD).<sup>1</sup> Although each of these conditions has a unique pathogenesis, they all are driven by a background of chronic inflammation. It has been reported that diets with high levels of refined carbohydrates and saturated or trans-fatty acids may exacerbate existing inflammation.<sup>2</sup> Consequently, dietary interventions, such as the ketogenic and low-glycemic diets, have potential anti-inflammatory and metabolic effects that are being assessed as stand-alone or adjunctive therapies for dermatologic diseases. </p> <p>Diet may partially influence systemic inflammation through its effect on weight. Higher body mass index and obesity are linked to a low-grade inflammatory state and higher levels of circulating inflammatory markers. Therefore, weight loss leads to decreases in inflammatory cytokines, including C-reactive protein, tumor necrosis factor <span class="body">α</span>, and IL-6.<sup>3</sup> These cytokines and metabolic effects overlap with inflammatory skin condition pathways. It also is posited that decreased insulin release associated with weight loss results in decreased sebaceous lipogenesis and androgens, which drive keratinocyte proliferation and acne development.<sup>4,5</sup> For instance, in a 2015 meta-analysis of 5 randomized controlled trials on psoriasis, patients in the weight loss intervention group had more substantial reductions in psoriasis area and severity index (PASI) scores compared with controls receiving usual care (<i>P</i><span class="body">=</span>.004).<sup>6</sup> However, in a systematic review of 35 studies on acne vulgaris, overweight and obese patients (defined by a body mass index of ≥23 kg/m<sup>2</sup>) had similar odds of having acne compared with normal-weight individuals (<i>P</i><span class="body">=</span>.671).<sup>7</sup> <br/><br/>Similar to weight loss, ketogenesis acts as a negative feedback mechanism to reduce insulin release, leading to decreased inflammation and androgens that often exacerbate inflammatory skin diseases.<sup>8</sup> Ketogenesis ensues when daily carbohydrate intake is limited to less than 50 g, and long-term adherence to a ketogenic diet results in metabolic reliance on ketone bodies such as acetoacetate, <span class="body">β</span>-hydroxybutyrate, and acetone.<sup>9</sup> These metabolites may decrease free radical damage and consequently improve signs and symptoms of acne, psoriasis, and other inflammatory skin diseases.<sup>10-12</sup> Similarly, increased ketones also may decrease activation of the NLRP3 (NOD-, LRR-, and Pyrin domain-containing protein 3) inflammasome and therefore reduce inflammatory markers such as IL-1<span class="body">β </span>and IL-1.<sup>4,13</sup> Several proposed mechanisms are outlined in the Table. <br/><br/>Collectively, low-glycemic and ketogenic diets have been proposed as potential interventions for reducing inflammatory skin conditions. These dietary approaches are hypothesized to exert their effects by facilitating weight loss, elevating ketone levels, and reducing systemic inflammation. The current review summarizes the existing evidence on ketogenic and low-glycemic diets as treatments for inflammatory skin conditions and evaluates the potential benefits of these dietary interventions in managing and improving outcomes for individuals with inflammatory skin conditions. </p> <h3>Methods</h3> <p>Using PubMed for articles indexed for MEDLINE and Google Scholar, a review of the literature was conducted with a combination of the following search terms: <i>low-glycemic diet, inflammatory, dermatologic, ketogenic diet, inflammation, dermatology, acne, psoriasis, eczema, seborrheic dermatitis</i>, and <i>hidradenitis suppurativa</i>. Reference citations in identified works also were reviewed. Interventional (experimental studies or clinical trials), survey-based, and observational studies that investigated the effects of low-glycemic or ketogenic diets for the treatment of inflammatory skin conditions were included. Inclusion criteria were studies assessing acne, psoriasis, SD, AD, and HS. Exclusion criteria were studies published before 1965; those written in languages other than English; and those analyzing other diets, such as the Mediterranean or low-fat diets. The search yielded a total of 11 observational studies and 4 controlled studies published between 1966 and January 2023. Because this analysis utilized publicly available data and did not qualify as human subject research, institutional review board approval was not required.</p> <h3>Results</h3> <p><i>Acne Vulgaris</i>—Acne vulgaris is a disease of chronic pilosebaceous inflammation and follicular epithelial proliferation associated with <i>Propionibacterium acnes</i>. The association between acne and low-glycemic diets has been examined in several studies. Diet quality is measured and assessed using the glycemic index (GI), which is the effect of a single food on postprandial blood glucose, and the glycemic load, which is the GI adjusted for carbohydrates per serving.<sup>14</sup> High levels of GI and glycemic load are associated with hyperinsulinemia and an increase in insulinlike growth factor 1 concentration that promotes <hl name="7"/>mechanistic target of rapamycin (mTOR) complex 1–mediated follicular lipogenesis, sebum fatty acid production, and androgen synthesis.<sup>15</sup> <i>Propionibacterium acnes </i>directly activates toll-like receptor 2 on monocytes through damage-associated molecular patterns and indirectly through products of triglyceride catalysis, causing release of IL-12, IL-6, tumor necrosis factor <span class="body">α</span>, and other proinflammatory cytokines.<sup>16</sup> Therefore, lifestyle modifications focused on strict glucose control have been postulated to reduce acne severity via modulation of lipogenesis, androgen concentration, and inflammation. </p> <p>Six survey-based studies evaluated sugar intake in patients with acne compared to healthy matched controls (eTable). Among these studies, 5 reported higher glycemic loads or daily sugar intake in acne patients compared to individuals without acne.<sup>12,19,20,26,28</sup> The remaining study was conducted in 1967 and enrolled 16 acne patients and 32 matched controls. It reported no significant difference in sugar intake between the groups (<i>P</i><span class="body">&gt;</span>.05).<sup>17<br/><br/></sup>Smith et al<sup>18</sup> randomized 43 male patients aged 15 to 25 years with facial acne into 2 cohorts for 12 weeks, each consuming either a low-glycemic diet (25% protein, 45% low-glycemic food [fruits, whole grains], and 30% fat) or a carbohydrate-dense diet of foods with medium to high GI based on prior documentation of the original diet. Patients were instructed to use a noncomedogenic cleanser as their only acne treatment. At 12 weeks, patients consuming the low-glycemic diet had an average of 23.5 fewer inflammatory lesions, while those in the intervention group had 12.0 fewer lesions (<i>P</i><span class="body">=</span>.03).<sup>18</sup> <br/><br/>In another controlled study by Kwon et al,<sup>21</sup> 32 male and female acne patients were randomized to a low-glycemic diet (25% protein, 45% low-glycemic food, and 30% fat) or a standard diet for 10 weeks. Patients on the low-glycemic diet experienced a 70.9% reduction in inflammatory lesions (<i>P</i><span class="body">&lt;</span>.05). Hematoxylin and eosin staining and image analysis were performed to measure sebaceous gland surface area in the low-glycemic diet group, which decreased from 0.32 to 0.24 mm<sup>2</sup> (<i>P</i><span class="body">=</span>.03). The sebaceous gland surface area in the control group was not reported. Moreover, patients on the low-glycemic diet had reduced IL-8 immunohistochemical staining (decreasing from 2.9 to 1.7 [<i>P</i><span class="body">=</span>.03]) and sterol regulatory element-binding protein 1 levels (decreasing from 2.6 to 1.3 [<i>P</i><span class="body">=</span>.03]), suggesting suppression of ongoing inflammation. Patients on the low-glycemic diet had no significant difference in transforming growth factor <span class="body">β</span>1(<i>P</i><span class="body">=</span>.83). In the control group, there was no difference in IL-8, sterol regulatory element binding protein 1, or transforming growth factor <span class="body">β</span>1 (<i>P</i><span class="body">&gt;</span>.05) on immunohistochemical staining.<sup>21<br/><br/></sup><i>Psoriasis</i>—Psoriasis is a systemic inflammatory disease characterized by hyperproliferation and aberrant keratinocyte plaque formation. The innate immune response of keratinocytes in response to epidermal damage or infection begins with neutrophil recruitment and dendritic cell activation. Dendritic cell secretion of IL-23 promotes T-cell differentiation into helper T cells (T<sub>H</sub>1) that subsequently secrete IL-17 and IL-22, thereby stimulating keratinocyte proliferation and eventual plaque formation. The relationship between diet and psoriasis is poorly understood; however, hyperinsulinemia is associated with greater severity of psoriasis.<sup>31</sup> <br/><br/>Four observational studies examined sugar intake in psoriasis patients. Barrea et al<sup>23</sup> conducted a survey-based study of 82 male participants (41 with psoriasis and 41 healthy controls), reporting that PASI score was correlated with intake of simple carbohydrates (percentage of total kilocalorie)(<em>r</em><span class="body">=</span>0.564, <i>P</i><span class="body">&lt;</span>.001). Another study by Yamashita et al<sup>27</sup> found higher sugar intake in psoriasis patients than controls (<i>P</i><span class="body">=</span>.003) based on surveys from 70 patients with psoriasis and 70 matched healthy controls.<br/><br/>These findings contrast with 2 survey-based studies by Johnson et al<sup>22</sup> and Afifi et al<sup>25</sup> of sugar intake in psoriasis patients using the National Health and Nutrition Examination Survey. Johnson et al<sup>22</sup> reported reduced sugar intake among 156 psoriasis patients compared with 6104 unmatched controls (odds ratio, 0.998; CI, 0.996-1 [<i>P</i><span class="body">=</span>.04]) from 2003 to 2006. Similarly, Afifi et al<sup>25</sup> reported decreased sugar intake in 1206 psoriasis patients compared with sex- and age-matched controls (<i>P</i><span class="body">&lt;</span>.0001) in 2009 and 2010. When patients were asked about dietary triggers, 13.8% of psoriasis patients reported sugar as the most common trigger, which was more frequent than alcohol (13.6%), gluten (7.2%), and dairy (6%).<sup>25<br/><br/></sup>Castaldo et al<sup>29,30</sup> published 2 nonrandomized clinical intervention studies in 2020 and 2021 evaluating the impact of the ketogenic diet on psoriasis. In the first study, 37 psoriasis patients followed a 10-week diet consisting of 4 weeks on a ketogenic diet (500 kcal/d) followed by 6 weeks on a low-caloric Mediterranean diet.<sup>29</sup> At the end of the intervention, there was a 17.4% reduction in PASI score, a 33.2-point reduction in itch severity score, and a 13.4-point reduction in the dermatology life quality index score; however, this study did not include a control diet group for comparison.<sup>29</sup> The second study included 30 psoriasis patients on a ketogenic diet and 30 control patients without psoriasis on a regular diet.<sup>30</sup> The ketogenic diet consisted of 400 to 500 g of vegetables, 20 to 30 g of fat, and a proportion of protein based on body weight with at least 12 g of whey protein and various amino acids. Patients on the ketogenic diet had significant reduction in PASI scores (value relative to clinical features, 1.4916 [<i>P</i><span class="body">=</span>.007]). Furthermore, concentrations of cytokines IL-2 (<i>P</i><span class="body">=</span>.04) and IL-1<span class="body">β</span> (<i>P</i><span class="body">=</span>.006) decreased following the ketogenic diet but were not measured in the control group.<sup>30</sup> <br/><br/><i>Seborrheic Dermatitis</i>—Seborrheic dermatitis is associated with overcolonization of <i>Malassezia</i> species near lipid-rich sebaceous glands. <em>Malassezia</em> hydrolyzes free fatty acids, yielding oleic acids and leading to T-cell release of IL-8 and IL-17.<sup>32</sup> Literature is sparse regarding how dietary modifications may play a role in disease severity. In a survey study, Bett et al<sup>17</sup> compared 16 SD patients to 1:2 matched controls (N<span class="body">=</span>29) to investigate the relationship between sugar consumption and presence of disease. Two control cohorts were selected, 1 from clinic patients diagnosed with verruca and 1 matched by age and sex from a survey-based study at a facility in London, England. Sugar intake was measured both in total grams per day and in “beverage sugar” per day, defined as sugar taken in tea and coffee. There was higher total sugar and higher beverage sugar intake among the SD group compared with both control groups (<i>P</i><span class="body">&lt;</span>.05).<sup>17<br/><br/></sup><i>Atopic Dermatitis</i>—Atopic dermatitis is a disease of epidermal barrier dysfunction and IgE-mediated allergic sensitization.<sup>33</sup> There are several mechanisms by which skin structure may be disrupted. It is well established that filaggrin mutations inhibit stratum corneum maturation and lamellar matrix deposition.<sup>34</sup> Upregulation of IL-4–, IL-13–, and IL-17–secreting T<sub>H</sub>2 cells also is associated with disruption of tight junctions and reduction of filaggrin.<sup>35,36</sup> Given that a T cell–mediated inflammatory response is involved in disease pathogenesis, glycemic control is hypothesized to have therapeutic potential.<br/><br/>Nosrati et al<sup>24</sup> surveyed 169 AD patients about their perceived dietary triggers through a 61-question survey based on the National Health and Nutrition Examination Survey. Respondents were queried about their perceptions and dietary changes, such as removal or addition of specific food groups and trial of specific diets. Overall, 16.5% of patients reported sugar being a trigger, making it the fourth most common among those surveyed and less common than dairy (24.8%), gluten (18.3%), and alcohol (17.1%).<sup>24</sup> <br/><br/><i>Hidradenitis Suppurativa</i>—Hidradenitis suppurativa is driven by hyperkeratosis, dilatation, and occlusion of pilosebaceous follicular ducts, whose eventual rupture evokes a local acute inflammatory response.<sup>37</sup> The inciting event for both acne and HS involves mTOR complex–mediated follicular hyperproliferation andinsulinlike growth factor 1 stimulation of androgen receptors in pilosebaceous glands. Given the similarities between the pathogenesis of acne and HS, it is hypothesized that lifestyle changes, including diet modification, may have a beneficial effect on HS.<sup>38-40</sup></p> <h3>Comment</h3> <p><i>Acne</i>—Overall, there is strong evidence supporting the efficacy of a low-glycemic diet in the treatment of acne. Notably, among the 6 observational studies identified, there was 1 conflicting study by Bett et al<sup>17</sup> that did not find a statistically significant difference in glucose intake between acne and control patients. However, this study included only 16 acne patients, whereas the other 5 observational studies included 32 to 2255 patients.<sup>17</sup> The strongest evidence supporting low-glycemic dietary interventions in acne treatment is from 2 rigorous randomized clinical trials by Kwon et al<sup>21</sup> and Smith et al.<sup>18</sup> These trials used intention-to-treat models and maintained consistency in gender, age, and acne treatment protocols across both control and treatment groups. To ensure compliance with dietary interventions, daily telephone calls, food logs, and 24-hour urea sampling were utilized. Acne outcomes were assessed by a dermatologist who remained blinded with well-defined outcome measures. An important limitation of these studies is the difficulty in attributing the observed results solely to reduced glucose intake, as low-glycemic diets often lead to other dietary changes, including reduced fat intake and increased nutrient consumption.<sup>18,21</sup></p> <p>A 2022 systematic review of acne by Meixiong et al<sup>41</sup> further reinforced the beneficial effects of low-glycemic diets in the management of acne patients. The group reviewed 6 interventional studies and 28 observational studies to investigate the relationship among acne, dairy, and glycemic content and found an association between decreased glucose and dairy on reduction of acne.<sup>41<br/><br/></sup>It is likely that the ketogenic diet, which limits glucose, would be beneficial for acne patients. There may be added benefit through elevated ketone bodies and substantially reduced insulin secretion. However, because there are no observational or interventional studies, further research is needed to draw firm conclusions regarding diet for acne treatment. A randomized clinical trial investigating the effects of the ketogenic diet compared to the low-glycemic diet compared to a regular diet would be valuable.<br/><br/><i>Psoriasis</i>—Among psoriasis studies, there was a lack of consensus regarding glucose intake and correlation with disease. Among the 4 observational studies, 2 reported increased glucose intake among psoriasis patients and 2 reported decreased glucose intake. It is plausible that the variability in studies is due to differences in sample size and diet heterogeneity among study populations. More specifically, Johnson et al<sup>22</sup> and Afifi et al<sup>25</sup> analyzed large sample sizes of 6260 and 2412 US participants, respectively, and found decreased sugar intake among psoriasis patients compared to controls. In comparison, Barrea et al<sup>23</sup> and Yamashita et al<sup>27</sup> analyzed substantially smaller and more specific populations consisting of 82 Italian and 140 Japanese participants, respectively; both reported increased glucose intake among psoriasis patients compared to controls. These seemingly antithetical results may be explained by regional dietary differences, with varying proportions of meats, vegetables, antioxidants, and vitamins.<br/><br/>Moreover, the variation among studies may be further explained by the high prevalence of comorbidities among psoriasis patients. In the study by Barrea et al,<sup>23</sup> psoriasis patients had higher fasting glucose (<i>P</i><span class="body">=</span>.004) and insulin (<i>P</i><span class="body">=</span>.022) levels than healthy patients. After adjusting for body mass index and metabolic syndrome, the correlation coefficient measuring the relationship between the PASI score and intake of simple carbohydrates changed from <em>r</em><span class="body">=</span>0.564 (<i>P</i><span class="body">&lt;</span>.001) to <em>r</em><span class="body">=</span>0.352 (<i>P</i><span class="body">=</span>.028). The confounding impact of these comorbidities was further highlighted by Yamashita et al,<sup>27</sup> who found statistically significant differences in glucose intake between psoriasis and healthy patients (<i>P</i><span class="body">=</span>.003). However, they reported diminished significance on additional subgroup analysis accounting for potential comorbidities (<i>P</i><span class="body">=</span>.994).<sup>27</sup> Johnson et al<sup>22</sup> and Afifi et al<sup>25</sup> did not account for comorbidities; therefore, the 4 observational study results must be interpreted cautiously.<br/><br/>The 2 randomized clinical trials by Castaldo et al<sup>29,30</sup> weakly suggest that a ketogenic diet may be beneficial for psoriasis patients. The studies have several notable limitations, including insufficient sample sizes and control groups. Thus, the decreased PASI scores reported in psoriasis patients on the ketogenic diets are challenging to interpret. Additionally, both studies placed patients on highly restrictive diets of 500 kcal/d for 4 weeks. The feasibility of recommending such a diet to patients in clinical practice is questionable. Diets of less than 500 kcal/d may be dangerous for patients with underlying comorbidities and are unlikely to serve as long-term solutions.<sup>23</sup> To contextualize our findings, a 2022 review by Chung et al<sup>42</sup> examined the impact of various diets—low-caloric, gluten-free, Mediterranean, Western, and ketogenic—on psoriasis and reported insufficient evidence to suggest a benefit to the ketogenic diet for psoriasis patients, though the Mediterranean diet may be well suited for psoriasis patients because of improved cardiovascular health and reduced mortality.<br/><br/><i>Seborrheic Dermatitis</i>—Sanders et al<sup>43</sup> found that patients with a high-fruit diet had lower odds of having SD, while those on a Western diet had higher odds of having SD. Although the study did not measure glycemic load, it is conceivable that the high glycemic load characteristic of the Western diet contributed to these findings.<sup>43</sup> However, no studies have investigated the direct link between low-glycemic or ketogenic diets and SD, leaving this area open for further study. <br/><br/><i>Atopic Dermatitis</i>—It has been hypothesized that mitigating T cell–mediated inflammation via glucose control may contribute to the improvement in AD.<sup>35,36</sup> However, in one study, 16.5% of AD patients self-identified sugar as a dietary trigger, ranking fourth among other dietary triggers.<sup>24</sup> Thus, the connection between glucose levels and AD warrants further exploration.<br/><br/><i>Hidradenitis Suppurativa</i>—Given the role of metabolic and hormonal influence in HS as well as the overlapping pathophysiology with acne, it is possible that low-glycemic and ketogenic diets may have a role in improving HS.<sup>38-40</sup> However, there is a gap in observation and controlled studies investigating the link between low-glycemic or ketogenic diets and HS.</p> <h3>Conclusion</h3> <p>Our analysis focused on interventional and observational research exploring the effects of low-glycemic and ketogenic diets on associations and treatment of inflammatory skin conditions. There is sufficient evidence to counsel acne patients on the benefits of a low-glycemic diet as an adjunctive treatment for acne. Currently, there is insufficient evidence to recommend a low-glycemic or ketogenic diet as a treatment for patients with any other inflammatory skin disease. Prospective and controlled clinical trials are needed to clarify the utility of dietary interventions for treating inflammatory skin conditions.</p> <h2>References</h2> <p class="reference"> 1. Pickett K, Loveman E, Kalita N, et al. Educational interventions to improve quality of life in people with chronic inflammatory skin diseases: systematic reviews of clinical effectiveness and cost-effectiveness. <i>Health Technol Assess</i>. 2015;19:1-176, v-vi.</p> <p class="reference"> 2. Giugliano D, Ceriello A, Esposito K. The effects of diet on inflammation: emphasis on the metabolic syndrome. <i>J Am Coll Cardiol</i>. 2006;48:677-685.<br/><br/> 3. Dowlatshahi EA, van der Voort EA, Arends LR, et al. Markers of systemic inflammation in psoriasis: a systematic review and meta-analysis. <i>Br J Dermatol</i>. 2013;169:266-282.<br/><br/> 4. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. <i>Nat Med</i>. 2015;21:263-269.<br/><br/> 5. Melnik BC. Acne vulgaris: the metabolic syndrome of the pilosebaceous follicle. <i>Clin Dermatol</i>. 2018;36:29-40.<br/><br/> 6. Upala S, Sanguankeo A. Effect of lifestyle weight loss intervention on disease severity in patients with psoriasis: a systematic review and meta-analysis. <i>Int J Obes (Lond)</i>. 2015;39:1197-1202.<br/><br/> 7. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. <i>Sci Rep</i>. 2020;10:5754.<br/><br/> 8. Paoli A, Grimaldi K, Toniolo L, et al. Nutrition and acne: therapeutic potential of ketogenic diets. <i>Skin Pharmacol Physiol</i>. 2012;25:111-117.<br/><br/> 9. Masood W, Annamaraju P, Khan Suheb MZ, et al. Ketogenic diet. <i>StatPearls</i>. StatPearls Publishing; 2023.<br/><br/>10. Fomin DA, McDaniel B, Crane J. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research. <i>J Dermatolog Treat</i>. 2017;28:484-487.<br/><br/>11. Zhang D, Jin W, Wu R, et al. High glucose intake exacerbates autoimmunity through reactive-oxygen-species-mediated TGF-<span class="body">β</span> cytokine activation. <i>Immunity</i>. 2019;51:671-681.e5.<br/><br/>12. Cerman AA, Aktas E, Altunay IK, et al. Dietary glycemic factors, insulin resistance, and adiponectin levels in acne vulgaris. <i>J Am Acad Dermatol</i>. 2016;75:155-162.<br/><br/>13. Ferrere G, Tidjani Alou M, Liu P, et al. Ketogenic diet and ketone bodies enhance the anticancer effects of PD-1 blockade. <i>JCI Insight</i>. 2021;6:e145207.<br/><br/>14. Burris J, Shikany JM, Rietkerk W, et al. A Low glycemic index and glycemic load diet decreases insulin-like growth factor-1 among adults with moderate and severe acne: a short-duration, 2-week randomized controlled trial. <i>J Acad Nutr Diet</i>. 2018;118:1874-1885.<br/><br/>15. Tan JKL, Stein Gold LF, Alexis AF, et al. Current concepts in acne pathogenesis: pathways to inflammation. <i>Semin Cutan Med Surg</i>. 2018;37(3S):S60-S62.<br/><br/>16. Kim J, Ochoa MT, Krutzik SR, et al. Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses.<i> J Immunol</i>. 2002;169:1535-1541.<br/><br/>17. Bett DG, Morland J, Yudkin J. Sugar consumption in acne vulgaris and seborrhoeic dermatitis. <i>Br Med J</i>. 1967;3:153-155.<br/><br/>18. Smith RN, Mann NJ, Braue A, et al. A low-glycemic-load diet improves symptoms in acne vulgaris patients: a randomized controlled trial. <i>Am J Clin Nutr</i>. 2007;86:107-115.<br/><br/>19. Rouhani P, Berman B, Rouhani G. Acne improves with a popular, low glycemic diet from South Beach. <i>J Am Acad Dermatol</i>. 2009;60(Suppl 1):AB14.<br/><br/>20. Aksu AE, Metintas S, Saracoglu ZN, et al. Acne: prevalence and relationship with dietary habits in Eskisehir, Turkey. <i>J Eur Acad Dermatol Venereol</i>. 2012;26:1503-1509.<br/><br/>21. Kwon HH, Yoon JY, Hong JS, et al. Clinical and histological effect of a low glycaemic load diet in treatment of acne vulgaris in Korean patients: a randomized, controlled trial. <i>Acta Derm Venereol</i>. 2012;92:241-246.<br/><br/>22. Johnson JA, Ma C, Kanada KN, et al. Diet and nutrition in psoriasis: analysis of the National Health and Nutrition Examination Survey (NHANES) in the United States. <i>J Eur Acad Dermatol Venereol</i>. 2014;28:327-332.<br/><br/>23. Barrea L, Macchia PE, Tarantino G, et al. Nutrition: a key environmental dietary factor in clinical severity and cardio-metabolic risk in psoriatic male patients evaluated by 7-day food-frequency questionnaire. <i>J Transl Med</i>. 2015;13:303.</p> <p class="reference">24. Nosrati A, Afifi L, Danesh MJ, et al. Dietary modifications in atopic dermatitis: patient-reported outcomes. <i>J Dermatolog Treat</i>. 2017;28:523-538.<br/><br/>25. Afifi L, Danesh MJ, Lee KM, et al. Dietary behaviors in psoriasis: patient-reported outcomes from a U.S. national survey. <i>Dermatol Ther (Heidelb)</i>. 2017;7:227-242.<br/><br/>26. Burris J, Rietkerk W, Shikany JM, et al. Differences in dietary glycemic load and hormones in New York City adults with no and moderate/severe acne. <i>J Acad Nutr Diet</i>. 2017;117:1375-1383.<br/><br/>27. Yamashita H, Morita T, Ito M, et al. Dietary habits in Japanese patients with psoriasis and psoriatic arthritis: low intake of meat in psoriasis and high intake of vitamin A in psoriatic arthritis. <i>J Dermatol</i>. 2019;46:759-769.<br/><br/>28. Marson J, Baldwin HE. 12761 Acne, twins, and glycemic index: a sweet pilot study of diet and dietary beliefs. <i>J Am Acad Dermatol</i>. 2020;83(Suppl):AB110.<br/><br/>29. Castaldo G, Rastrelli L, Galdo G, et al. Aggressive weight-loss program with a ketogenic induction phase for the treatment of chronic plaque psoriasis: a proof-of-concept, single-arm, open-label clinical trial. <i>Nutrition</i>. 2020;74:110757.<br/><br/>30. Castaldo G, Pagano I, Grimaldi M, et al. Effect of very-low-calorie ketogenic diet on psoriasis patients: a nuclear magnetic resonance-based metabolomic study. <i>J Proteome Res</i>. 2021;20:1509-1521.<br/><br/>31. Ip W, Kirchhof MG. Glycemic control in the treatment of psoriasis. <i>Dermatology</i>. 2017;233:23-29.<br/><br/>32. Vijaya Chandra SH, Srinivas R, Dawson TL Jr, et al. Cutaneous <i>Malassezia</i>: commensal, pathogen, or protector? <i>Front Cell Infect Microbiol</i>. 2020;10:614446.<br/><br/>33. David Boothe W, Tarbox JA, Tarbox MB. Atopic dermatitis: pathophysiology. <i>Adv Exp Med Biol</i>. 2017;1027:21-37.<br/><br/>34. Guttman-Yassky E, Hanifin JM, Boguniewicz M, et al. The role of phosphodiesterase 4 in the pathophysiology of atopic dermatitis and the perspective for its inhibition. <i>Exp Dermatol</i>. 2019;28:3-10.<br/><br/>35. Furue K, Ito T, Tsuji G, et al. The IL-13–OVOL1–FLG axis in atopic dermatitis. Immunology. 2019;158:281-286.<br/><br/>36. Renert-Yuval Y, Guttman-Yassky E. New treatments for atopic dermatitis targeting beyond IL-4/IL-13 cytokines. <i>Ann Allergy Asthma Immunol</i>. 2020;124:28-35.<br/><br/>37. Sellheyer K, Krahl D. “Hidradenitis suppurativa” is acne inversa! An appeal to (finally) abandon a misnomer. <i>Int J Dermatol</i>. 2005;44:535-540.<br/><br/>38. Danby FW, Margesson LJ. Hidradenitis suppurativa. <i>Dermatol Clin</i>. 2010;28:779-793.<br/><br/>39. Fernandez JM, Marr KD, Hendricks AJ, et al. Alleviating and exacerbating foods in hidradenitis suppurativa. <i>Dermatol Ther</i>. 2020;33:E14246.<br/><br/>40. Yamanaka-Takaichi M, Revankar R, Shih T, et al. Expert consensus on priority research gaps in dietary and lifestyle factors in hidradenitis suppurativa: a Delphi consensus study. <i>Arch Dermatol Res</i>. 2023;315:2129-2136.<br/><br/>41. Meixiong J, Ricco C, Vasavda C, et al. Diet and acne: a systematic review. <i>JAAD Int</i>. 2022;7:95-112.<br/><br/>42. Chung M, Bartholomew E, Yeroushalmi S, et al. Dietary intervention and supplements in the management of psoriasis: current perspectives. <i>Psoriasis (Auckland)</i>. 2022;12:151-176. <span class="citation-doi">doi:10.2147/PTT.S328581<br/><br/></span>43. Sanders MGH, Pardo LM, Ginger RS, et al. Association between diet and seborrheic dermatitis: a cross-sectional study. <i>J Invest Dermatol</i>. 2019;139:108-114.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Katie Roster, Lillian Xie, and Terry Nguyen are from New York Medical College, Valhalla. Dr. Lipner is from the Department of Dermatology,Weill Cornell Medicine, New York, New York.</p> <p class="disclosure">Katie Roster, Lillian Xie, and Terry Nguyen report no conflict of interest. Dr. Lipner has been a consultant for Ortho Dermatologics; has received research grants from BelleTorus Corporation and Moberg Pharma; and has served on the board for Hoth Therapeutics.<br/><br/>The eTable is available in the Appendix online at www.mdedge.com/dermatology.<br/><br/>Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, 9th Floor, New York, NY 10021 (shl9032@med.cornell.edu).<br/><br/>doi:10.12788/cutis.0942</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>As the ketogenic diet gains in popularity, dermatologists may inform patients that there is emerging evidence supporting the idea that low-glycemic diets may contribute to improvement in inflammatory skin conditions. </li> <li>Dermatologists may educate patients about the potential benefits of a low-glycemic diet as a supplementary treatment for acne based on existing evidence. </li> <li>Current evidence is insufficient to endorse a ketogenic diet as superior to other dietary approaches in treating inflammatory skin conditions. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • As the ketogenic diet gains in popularity, dermatologists may inform patients that there is emerging evidence supporting the idea that low-glycemic diets may contribute to improvement in inflammatory skin conditions.
  • Dermatologists may educate patients about the potential benefits of a low-glycemic diet as a supplementary treatment for acne based on existing evidence.
  • Current evidence is insufficient to endorse a ketogenic diet as superior to other dietary approaches in treating inflammatory skin conditions.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026B5.SIG
Disable zoom
Off

Expanding the Psoriasis Framework: Immunopathogenesis and Treatment Updates

Article Type
Changed
Mon, 02/05/2024 - 10:00
Display Headline
Expanding the Psoriasis Framework: Immunopathogenesis and Treatment Updates

Psoriasis is a chronic inflammatory disease that affects approximately 3% of the US population.1 Plaque psoriasis comprises 80% to 90% of cases, while pustular, erythrodermic, guttate, inverse, and palmoplantar disease are less common variants (Figure 1). Psoriatic skin manifestations range from localized to widespread or generalized disease with recurrent flares. Body surface area or psoriasis area and severity index (PASI) measurements primarily focus on skin manifestations and are important for evaluating disease activity and response to treatment, but they have inherent limitations: they do not capture extracutaneous disease activity, systemic inflammation, comorbid conditions, quality of life impact, or the economic burden of psoriasis.

CT11302082_ABCD.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Characteristic%20plaque%20psoriasis%20of%20the%20trunk.%20C%2C%20Inverse%20psoriasis%20involving%20the%20inframammary%20folds.%20D%2C%20Guttate%20psoriasis%20in%20an%20adult%20following%20streptococcal%20infection.%3C%2Fp%3E

A common manifestation of psoriasis is psoriatic arthritis (PsA), which can involve the nails, joints, ligaments, or tendons in 30% to 41% of affected individuals (Figure 2).2,3 A growing number of psoriasis-associated comorbidities also have been reported including metabolic syndrome4; hyperlipidemia5; cardiovascular disease6; stroke7; hypertension8; obesity9; sleep disorders10; malignancy11; infections12; inflammatory bowel disease13; and mental health disorders such as depression,14 anxiety,15 and suicidal ideation.15 Psoriatic disease also interferes with daily life activities and a patient’s overall quality of life, including interpersonal relationships, intimacy, employment, and work productivity.16 Finally, the total estimated cost of psoriasis-related health care is more than $35 billion annually,17 representing a substantial economic burden to our health care system and individual patients.

Nong_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Clinical%20manifestation%20of%20psoriatic%20arthritis%20involving%20the%20metacarpal%20joints%20of%20the%20hands.%3C%2Fp%3E

The overall burden of psoriatic disease has declined markedly in the last 2 decades due to revolutionary advances in our understanding of the immunopathogenesis of psoriasis and the subsequent development of improved therapies that predominantly interrupt IL-23/IL-17 cytokine signaling; however, critical knowledge and treatment gaps persist, underscoring the importance of ongoing clinical and research efforts in psoriatic disease. We review the working immune model of psoriasis, summarize related immune discoveries, and highlight recent therapeutic innovations that are shaping psoriatic disease management.

Current Immune Model of Psoriatic Disease

Psoriasis is an autoinflammatory T cell–mediated disease with negligible contributions from the humoral immune response. Early clinical observations reported increased inflammatory infiltrates in psoriatic skin lesions primarily consisting of both CD4+ and CD8+ T-cell populations.18,19 Additionally, patients treated with broad-acting, systemic immunosuppressive medications (eg, cyclosporine, oral corticosteroids) experienced improvement of psoriatic lesions and normalization of the immune infiltrates observed in skin biopsy specimens.20,21 These early clinical findings led to more sophisticated experimentation in xenotransplant models of psoriasis,22,23 which explored the clinical efficacy of several less immunosuppressive (eg, methotrexate, anti–tumor necrosis factor [TNF] biologics)24 or T cell–specific agents (eg, alefacept, abatacept, efalizumab).25-27 The results of these translational studies provided indisputable evidence for the role of the dysregulated immune response as the primary pathogenic process driving plaque formation; they also led to a paradigm shift in how the immunopathogenesis of psoriatic disease was viewed and paved the way for the identification and targeting of other specific proinflammatory signals produced by activated dendritic cell (DC) and T-lymphocyte populations. Among the psoriasis-associated cytokines subsequently identified and studied, elevated IL-23 and IL-17 cytokine levels in psoriatic skin were most closely associated with disease activity, and rapid normalization of IL-23/IL-17 signaling in response to effective oral or injectable antipsoriatic treatments was the hallmark of skin clearance.28 The predominant role of IL-23/IL-17 signaling in the development and maintenance of psoriatic disease is the central feature of all working immune models for this disease (Figure 3).

Nong_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Working%20immune%20model%20of%20psoriasis.%20Early%20immune%20events%20include%20activation%20of%20dendritic%20cells%20(DCs)%20and%20IL-17%E2%80%93producing%20T%20cells%20(T17)%20in%20the%20prepsoriatic%20(or%20normal-appearing)%20skin%20of%20individuals%20who%20are%20genetically%20susceptible%20and%2For%20have%20exposures%20to%20known%20psoriasis%20triggers.%20Activation%20of%20DC%20and%20T17%20populations%20in%20the%20skin%20results%20in%20increased%20production%20of%20tumor%20necrosis%20factor%20(TNF)%2C%20IL-23%2C%20and%20IL-17%20cytokines%20(namely%20IL-17A%20and%20IL-17F)%2C%20which%20work%20synergistically%20with%20other%20immune%20signals%20(IL-12%2C%20IL-22%2C%20IL-36%2C%20TNF%2C%20interferon%20%5BIFN%5D)%20to%20drive%20keratinocyte%20(KC)%20hyperproliferation.%20In%20response%20to%20upregulated%20IL-17%20signaling%2C%20substantial%20increases%20in%20keratinocyte-derived%20proteins%20(antimicrobial%20peptides%2C%20IL-19%2C%20IL-36%2C%20IL-17C)%20and%20chemotactic%20factors%20(chemokine%20%5BC-C%20motif%5D%20ligand%2020%20%5BCCL20%5D%2C%20chemokine%20%5BC-C%20motif%5D%20ligand%201%2F2%2F3%2F5%2F8%20%5BCXCL1%2F2%2F3%2F5%2F8%5D%5Bor%20IL-8%5D)%20facilitate%20further%20activation%20and%20recruitment%20of%20T17%20and%20helper%20T%20cell%20(TH1)%20lymphocytes%2C%20DCs%2C%20macrophages%2C%20and%20polymorphonuclear%20neutrophils%20(PMNs)%20into%20the%20skin.%20The%20resultant%20inflammatory%20circuit%20creates%20a%20self-amplifying%20or%20feed-forward%20immune%20response%20in%20the%20skin%20that%20leads%20to%20the%20hallmark%20clinical%20features%20of%20psoriasis%20and%20sustains%20the%20mature%20psoriatic%20plaque.%3C%2Fp%3E

Psoriasis-Associated Genetic and Environmental Risk Factors

The exact sequence of events that lead to the initiation and formation of plaque psoriasis in susceptible individuals is still poorly understood; however, several important risk factors and key immune events have been identified. First, decades of genetic research have reported more than 80 known psoriasis-associated susceptibility loci,29 which explains approximately 50% of psoriasis heritability. The major genetic determinant of psoriasis, HLA-C*06:02 (formerly HLA-Cw6), resides in the major histocompatibility complex class I region on chromosome 6p21.3 (psoriasis susceptibility gene 1, PSORS1) and is most strongly associated with psoriatic disease.30 Less common psoriasis-associated susceptibility genes also are known to directly or indirectly impact innate and adaptive immune functions that contribute to the pathogenesis of psoriasis.

Second, several nongenetic environmental risk factors for psoriasis have been reported across diverse patient populations, including skin trauma/injury, infections, alcohol/tobacco use, obesity, medication exposure (eg, lithium, antimalarials, beta-blockers), and stress.31 These genetic and/or environmental risk factors can trigger the onset of psoriatic disease at any stage of life, though most patients develop disease in early adulthood or later (age range, 50–60 years). Some patients never develop psoriasis despite exposure to environmental risk factors and/or a genetic makeup that is similar to affected first-degree relatives, which requires further study.

Prepsoriatic Skin and Initiation of Plaque Development

In response to environmental stimuli and/or other triggers of the immune system, DC and resident IL-17–producing T-cell (T17) populations become activated in predisposed individuals. Dendritic cell activation leads to the upregulation and increase of several proinflammatory cytokines, including TNF, interferon (IFN) α, IFN-γ, IL-12, and IL-23. Tumor necrosis factor and IL-23 play a vital role in psoriasis by helping to regulate the polarization and expansion of T22 and T17 cells in the skin, whereas IL-12 promotes a corresponding type 1 inflammatory response.32 Increased IL-17 and IL-22 result in alteration of the terminal differentiation and proliferative potential of epidermal keratinocytes, leading to the early clinical hallmarks of psoriatic plaques. The potential contribution of overexpressed psoriasis-related autoantigens, such as LL-37/cathelicidin, ADAMTSL5, and PLA2G4D,33 in the initiation of psoriatic plaques has been suggested but is poorly characterized.34 Whether these specific autoantigens or others presented by HLA-C variants found on antigen-presenting cells are required for the breakdown of immune tolerance and psoriatic disease initiation is highly relevant but requires further investigation and validation.

 

 

Feed-Forward Inflammation, Mature Psoriatic Plaques, and Resident Memory T Cells

In response to the upstream production of IL-23 by dermal DCs, high levels of IL-17 cytokines can be found in mature psoriatic plaques. The IL-17 family consists of 6 dimeric cytokines (IL-17A through IL-17F) that provide innate cutaneous protection against bacterial, viral, and fungal infectious agents, such as Candida albicans. Unlike other IL-17 isoforms, IL-17A and IL-17F share the same receptor complex and have the highest structural homology of any pair (approximately 50% similar).35 The relative expression of IL-17F is higher than IL-17A in psoriasis,36 though IL-17A has been considered as the predominant IL-17 cytokine found in psoriatic skin lesions due to its higher potency.

Binding of IL-17A/F with the IL-17 receptor (IL-17R) on keratinocytes contributes to the development of psoriatic plaques by inducing epidermal hyperplasia via activation of CCAAT/enhancer-binding proteins β and δ, nuclear factor κB, and signal transducer and activator of transcription 1 gene (STAT1).37,38 This also increases the expression of other keratinocyte-derived proteins (eg, human β-defensins, S-100 proteins, LL-37, other antimicrobial peptides, IL-19, IL-36, IL-17C) that act as reinforcing proinflammatory signals or chemotactic factors (eg, chemokine [C-C motif] ligand 20 [CCL20], chemokine [C-C motif] ligand 1/2/3/5 [CXCL1/2/3/5], CXCL8, IL-8) that facilitate the recruitment of additional immune cells to the skin including polymorphonuclear neutrophils (PMNs), macrophages, and DCs.39-41 Routine immunohistochemical staining for these keratinocyte-derived proteins reveals a striking epidermal gene expression gradient wherein levels of IL-17–induced proteins are most highly expressed in the uppermost layers of keratinocytes and facilitate the recruitment of immune cells into the epidermis. Activated T17 cells also stimulate the production of keratinocyte-derived chemokines (eg, CXCL9/10/11), which recruit type 1 inflammatory T-cell populations into developing psoriatic plaques.42,43 Finally, TNF, IL-36, and IL-17C cytokines act synergistically with IL-17A/F to amplify the proinflammatory effects of IL-17 signaling and further stimulate their production from T17 cell populations.40 This inflammatory circuit in the skin creates and supports a self-amplifying or positive feedback loop between the skin and immune system that commonly is referred to as feed-forward inflammation (Figure 3).34 The feed-forward inflammatory loop in psoriasis—predominantly driven by increased IL-23/IL-17 signaling—best characterizes the mature psoriatic plaque.

Several findings suggest that the influx of persistent, long-lived resident memory T cells (Trms) may contribute to the mature psoriatic plaque. It is believed that CD8+CD103+CD49a Trm cell populations may be responsible for the sharply demarcated borders of untreated psoriasis plaques or their recurrence at specific body sites such as the scalp, buttocks, extremity extensor surfaces, umbilicus, or acral skin following specific stimuli or trauma (Koebner phenomenon or isomorphic response).44,45 It is not known if repeated stimuli or trauma induce disease formation via the activation of Trm cell populations; further study in large patient cohorts is needed, but this remains an intriguing area of study for durable treatment responses and potential cures for psoriasis.

Recent Discoveries in Psoriatic Disease

Remarkable treatment outcomes for psoriasis have been achieved with multiple selective IL-17 and IL-23 inhibitors (eTable). As demonstrated in several pivotal phase 3 clinical trials for members of these classes of medications, the majority of treated psoriasis patients achieved PASI90 clearance.46 Due to their more favorable dosing schedule (ie, fewer injections) and ability to induce a durable remissionlike treatment response, IL-23 inhibitors have become the preferred treatment class for cutaneous disease, while IL-17 inhibitors may be preferred when treating patients with both plaque psoriasis and PsA.47,48 Nevertheless, the complexity of this disease is punctuated by treated patients who do not adequately respond to selective IL-23/IL-17 blockade.49 Recent and emerging treatments may shed light on these recalcitrant cases and will add to the rapidly growing arsenal of available psoriasis therapies.

The Role of IL-17F in Psoriasis and Other Inflammatory Skin Diseases

Dysregulation of IL-17A and IL-17F is associated with several chronic inflammatory conditions, such as psoriasis and PsA.35,50 Both cytokines, either as homodimers or heterodimers, can selectively bind to the heterodimeric IL-17R formed by the IL-17RA and IL-17RC subunits.35 IL-17F and IL-17C also can synergize with TNF and other cytokines to promote and support the self-sustaining inflammatory circuits in mature psoriatic plaques, though their inflammatory effects in the skin are more limited than IL-17A.51,52 Therefore, incomplete blockade of IL-17 signaling (ie, unopposed IL-17F and IL-17C) represents a potential mechanism to explain the persistence of psoriasis in patients treated with selective IL-17A inhibitors. This hypothesis is supported by reports of psoriasis patients who have inadequate clinical responses to selective IL-17A inhibition but subsequently improve with IL-17R blockade, which results in disruption of IL-17A as well as IL-17C/E/F cytokine signaling. This formed the basis for further study into the specific role of IL-17F in psoriatic disease and any potential therapeutic benefits associated with its inhibition.

Recently approved in the European Union, Canada, Australia, Japan, the United Kingdom, and the United States for moderate to severe psoriasis, bimekizumab is a novel humanized IgG antibody that selectively inhibits both IL-17A and IL-17F cytokines.53 Specifically, bimekizumab simultaneously prevents binding of IL-17A/A, IL-17A/F, and IL-17F/F dimers with the IL-17R. Compared to other IL-17 and IL-23 biologic therapies, bimekizumab (320 mg) achieved relatively higher response rates for PASI75, PASI90, and PASI100.49 Neutralization of IL-17A and IL-17F by bimekizumab also resulted in more complete suppression of cytokine responses and PMN chemotaxis than either cytokine alone in treated PsA patients,54 which is notable because of the incremental benefits of recent IL-23 and IL-17 inhibitors on inflammatory arthritis symptoms in contrast to the substantial improvements observed for cutaneous disease with those same agents.

The primary disadvantage of bimekizumab and its more complete blockade of the IL-17 signaling pathway is that treated patients have a substantially increased risk for oral candidiasis (>10%).55 However, the precise link between candidiasis and IL-17 blockade is not yet fully understood because other targeted agents that also broadly suppress IL-17 signaling (ie, IL-17R, IL-23 inhibitors) are associated with much lower rates of candidiasis.56-58 Bimekizumab also is being investigated as a novel therapy for hidradenitis suppurativa and will provide important reference information regarding the role for bispecific biologic agents in the treatment of chronic inflammatory skin diseases.59

 

 

IL-36 Signaling and Generalized Pustular Psoriasis

Recent genetic and clinical studies have expanded our understanding of the role of IL-36 signaling in the immunopathogenesis of pustular psoriasis variants. Generalized pustular psoriasis (GPP) is a rare distinct psoriasis subtype characterized by the recurrent development of widespread erythema, superficial sterile pustules, and desquamation. Systemic symptoms such as fever, malaise, itching, and skin pain accompany acute GPP flares.60 Generalized pustular psoriasis is more common in female patients (in contrast with plaque psoriasis), and acute flares may be caused by multiple stimuli including infections, hypocalcemia, initiation or discontinuation of medications (eg, oral corticosteroids), pregnancy, or stress.61,62 Flares of GPP often require emergency or in-patient care, as untreated symptoms increase the risk for severe health complications such as secondary infections, sepsis, or multisystem organ failure.63 The prevalence of GPP is estimated to be approximately 1 in 10,000 individuals in the United States,64-67 with mortality rates ranging from 0 to 3.3 deaths per 100 patient-years.67

In contrast to plaque psoriasis, aberrant IL-36 signaling is the predominant driver of GPP. IL-36 is a member of the IL-1 cytokine family that includes three IL-36 agonists (IL-36α, IL-36β, IL-36γ) and 1 endogenous antagonist (IL-36Ra, encoded by IL36RN).68 The immunopathogenesis of GPP involves dysregulation of the IL-36–chemokine–PMN axis, resulting in unopposed IL-36 signaling and the subsequent recruitment and influx of PMNs into the epidermis. IL36RN mutations are strongly associated with GPP and result in impaired function of the IL-36Ra protein, leading to unopposed IL-36 signaling.69 However, approximately two-thirds of GPP patients lack identifiable gene mutations, suggesting other immune mechanisms or triggers causing upregulated IL-36 signaling.70 In response to these triggers, increased IL-36 cytokines released by keratinocytes bind to the IL-36R, resulting in substantial keratinocyte hyperproliferation, increased IL-36 levels, and the expression of hundreds of additional inflammatory signals (eg, IL-17C, antimicrobial peptides, TNF, IL-6).71 Increased IL-36 levels also drive the production of PMN chemotactic proteins (eg, CXCL1/2/3/5/6/8 and CXCR1/2) and act synergistically with IL-17 cytokines to create an autoamplifying circuit that is analogous to the feed-forward inflammatory loop in plaque psoriasis.72 Biopsies of involved GPP skin reveal increased expression of IL-36 in the uppermost layers of the epidermis, which creates a gene expression gradient that acts as a strong attractant for PMNs and forms the basis for the hallmark pustular lesions observed in GPP patients.

Until recently, treatment strategies for GPP involved the off-label use of topical, oral, or biologic therapies approved for plaque psoriasis, which often was associated with variable or incomplete disease control. In September 2022, the US Food and Drug Administration (FDA) approved intravenous spesolimab as a first-in-class humanized monoclonal IgG1 antibody for the treatment of GPP flares in adults. Spesolimab binds to IL-36R and prevents its activation by its endogenous agonists. A phase 2, randomized, 12-week clinical trial (Effisayil-1) evaluated the efficacy and safety of a single 900-mg intravenous dose of spesolimab followed by an optional second dose 1 week later for inadequate treatment responses in 53 enrolled GPP patients (2:1 treatment to placebo randomization).73 Remarkably, more than half (19/35 [54%]) of GPP patients experienced complete resolution of pustules (GPP physician global assessment subscore of 0 [range, 0–4]) and showed sustained efficacy out to week 12 after just 1 or 2 doses of spesolimab. Overall, the safety profile of spesolimab was good; asthenia, fatigue, nausea, vomiting, headache, pruritus, infusion-related reaction and symptoms, and mild infections (eg, urinary tract infection) were the most common adverse events reported.73

Imsidolimab, a high-affinity humanized IgG4 monoclonal antibody that binds and blocks activation of IL-36R, also has completed phase 2 testing,74 with phase 3 study results expected in early 2024. The rapid onset of action and overall safety of imsidolimab was in line with and similar to spesolimab. Future approval of imsidolimab would add to the limited treatment options available for GPP and has the additional convenience of being administered to patients subcutaneously. Overall, the development of selective IL-36R inhibitors offers a much-needed therapeutic option for GPP and illustrates the importance of translational research.

Role of Tyrosine Kinase in Psoriatic Disease

The Janus kinase (JAK) enzyme family consists of 4 enzymes—tyrosine kinase 2 (TYK2), JAK1, JAK2, and JAK3—that function as intracellular transduction signals that mediate the biologic response of most extracellular cytokines and growth factors.75 Critical psoriasis-related cytokines are dependent on intact JAK-STAT signaling, including IL-23, IL-12, and type I IFNs. In 2010, a genome-wide association identified TYK2 as a psoriasis susceptibility locus,76 and loss-of-function TYK2 mutations confer a reduced risk for psoriasis.77 Unlike other JAK isoforms, TYK2 mediates biologic functions that are highly restricted to the immune responses associated with IL-23, IL-12, and type I IFN signaling.78,79 For these reasons, blockade of TYK2 signaling is an attractive therapeutic target for the potential treatment of psoriatic disease.

In September 2022, the FDA approved deucravacitinib as a first-in-class, oral, selective TYK2 inhibitor for the treatment of adult patients with moderate to severe plaque psoriasis. It was the first FDA approval of an oral small-molecule treatment for plaque psoriasis in nearly a decade. Deucravacitinib inhibits TYK2 signaling via selective binding of its unique regulatory domain, resulting in a conformational (allosteric) change that interferes with its active domain.80 This novel mechanism of action limits the unwanted blockade of other broad biologic processes mediated by JAK1/2/3. Of note, the FDA did not issue any boxed warnings for deucravacitinib as it did for other FDA-approved JAK inhibitors.

In a head-to-head, 52-week, double-blind, prospective, randomized, phase 3 study, deucravacitinib showed clear superiority over apremilast for PASI75 at week 16 (53.0% [271/511] vs 39.8% [101/254]) and week 24 (58.7% [296/504] vs 37.8% [96/254]).81 Clinical responses were sustained through week 52 and showed efficacy for difficult-to-treat areas such as the scalp, acral sites, and nails. Other advantages of deucravacitinib include once-daily dosing with no need for dose titration or adjustments for renal insufficiency as well as the absence of statistically significant differences in gastrointestinal tract symptoms compared to placebo. The most common adverse effects included nasopharyngitis, upper respiratory tract infections, headache, diarrhea, and herpes infections.81 The potential benefit of deucravacitinib for PsA and psoriasis comorbidities remains to be seen, but it is promising due to its simultaneous disruption of multiple psoriasis-related cytokine networks. Several other TYK2 inhibitors are being developed for psoriatic disease and related inflammatory conditions, underscoring the promise of targeting this intracellular pathway.

 

 

Aryl Hydrocarbon Receptor Agonism

Topical steroids are the mainstay treatment option for localized or limited plaque psoriasis due to their potent immunosuppressive effect on the skin and relatively low cost. Combined with vitamin D analogs, topical steroids result in marked improvements in disease severity and improved tolerability.82 However, chronic use of topical steroids is limited by the need for twice-daily application, resulting in poor treatment compliance; loss of efficacy over time; risk for steroid-induced skin atrophy on special body sites; and patient concerns of potential systemic effects. The discovery of novel drug targets amenable to topical inhibition is needed.

Dysregulated aryl hydrocarbon receptor (AHR) levels have been reported in atopic dermatitis and psoriasis.83 Aryl hydrocarbon receptors are ubiquitously expressed in many cell types and play an integral role in immune homeostasis within the skin, skin barrier function, protection against oxidative stressors, and regulation of proliferating melanocytes and keratinocytes.84,85 They are widely expressed in multiple immune cell types (eg, antigen-presenting cells, T lymphocytes, fibroblasts) and modulate the differentiation of T17 and T22 cells as well as their balance with regulatory T-cell populations.86 In keratinocytes, AHR helps to regulate terminal differentiation, enhance skin barrier integrity via AHR-dependent filaggrin (FLG) expression, and prevent transepidermal water loss.87,88 The mechanisms by which AHR ligands lead to the upregulation or downregulation of specific genes is intricate and highly context dependent, such as the specific ligand and cell type involved. In preclinical studies, AHR-deficient mice develop psoriasiform skin inflammation, increased IL-17 and IL-22 expression, and abnormal skin barrier function.89 Keratinocytes treated with AHR ligands in vitro modulated psoriasis-associated inflammatory cytokines, such as IL-6, IL-8, and type I and II IFNs.89,90 The use of coal tar, one of the earliest historical treatments for psoriasis, is thought to activate AHRs in the skin via organic compound mixtures containing polyaromatic hydrocarbons that help normalize the proinflammatory environment in psoriatic skin.91

In June 2022, the FDA approved tapinarof as a first-in-class, topical, nonsteroidal AHR agonist for the treatment of plaque psoriasis in adults. Although the exact mechanism of action for tapinarof has not been fully elucidated, early studies suggest that its primary function is the activation of AHR, leading to reduced T-cell expansion and T17 cell differentiation. In the imiquimod mouse model, cytokine expression of IL-17A, IL-17F, IL-19, IL-22, IL-23A, and IL-lβ in psoriasiform skin lesions were downregulated following tapinarof treatment.92 In humans, tapinarof treatment is associated with a remittive effect, in which the average time for tapinarof-treated psoriasis lesions to remain clear was approximately 4 months.93 Preliminary research investigating the mechanism by which tapinarof induces this remittive effect is ongoing and may involve the reduced activation and influx of T17 and Trm populations into the skin.94 However, these preclinical studies were performed on healthy dermatome-derived skin tissue cultured in T17-skewing conditions and needs to be replicated in larger samples sizes using human-derived psoriatic tissue. Alternatively, a strong inhibitory effect on IL-23 cytokine signaling may, in part, explain the remittive effect of tapinarof, as an analogous response is observed in patients who start and discontinue treatment with selective IL-23 antagonists. Regardless, the once-daily dosing of tapinarof and sustained treatment response is appealing to psoriasis patients. Tapinarof generally is well tolerated with mild folliculitis (>20% of patients) and contact dermatitis (5% of patients) reported as the most common skin-related adverse events.

New Roles for Phosphodiesterase 4 Inhibition

Phosphodiesterases (PDEs) are enzymes that hydrolyze cyclic nucleotides (eg, cyclic adenosine monophosphate) to regulate intracellular secondary messengers involved in the inflammatory response. One of several enzymes in the PDE family, PDE4, has been shown to have greater activity in psoriatic skin compared to healthy skin.95 Phosphodiesterase inhibitors decrease the degradation of cyclic adenosine monophosphate, which triggers protein kinase A to downregulate proinflammatory (eg, TNF-α, IL-6, IL-17, IL-12, IL-23) cytokines and increased expression of anti-inflammatory signals such as IL-10.96,97 Apremilast, the first oral PDE4 inhibitor approved by the FDA for psoriasis, offered a safe alternative to traditional oral immunosuppressive agents that had extensive risks and potential end-organ adverse effects. Unfortunately, apremilast demonstrated modest efficacy for psoriatic disease (better efficacy in the skin vs joint manifestations) and was supplanted easily by next-generation targeted biologic agents that were more efficacious and lacked the troublesome gastrointestinal tract adverse effects of PDE4 inhibition.98

Crisaborole became the first topical PDE4 inhibitor approved in the United States in December 2016 for twice-daily treatment of atopic dermatitis. Although phase 2 trial results were reported in psoriasis, this indication was never pursued, presumably due to similar improvements in primary outcome measures at week 12, compared to placebo (ClinicalTrials.gov Identifier NCT01300052).

In July 2022, the first topical PDE4 inhibitor indicated for plaque psoriasis was approved by the FDA—­roflumilast cream 0.3% for once-daily use in individuals 12 years and older. Roflumilast was found to be clinically efficacious as early as 2 weeks after its use in an early-phase clinical trial.99 In 2 phase 3 clinical trials (DERMIS-1 and DERMIS-2), roflumilast significantly increased the proportion of patients achieving PASI75 at week 8 compared to vehicle (39%–41.6% vs 5.3%–7.6%, respectively)(P<.001).100 Overall, this nonsteroidal topical therapy was found to be well tolerated, with infrequent reports of application site pain or irritation as adverse events. Similar to tapinarof, patients can apply roflumilast on all body surface areas including the face, external genitalia, and other intertriginous areas.100 Importantly, the broad immune impact of PDE4 inhibition suggests that topical roflumilast likely will be an effective treatment for several additional inflammatory conditions, including seborrheic dermatitis and atopic dermatitis, which would expand the clinical utility of this specific medication.

Conclusion

In the last 2 decades, we have witnessed a translational revolution in our understanding of the underlying genetics and immunology of psoriatic disease. Psoriasis is widely considered one of the best-managed inflammatory conditions in all of medicine due to the development and availability of highly targeted, effective topical and systemic therapies that predominantly disrupt IL-23/IL-17 cytokine signaling in affected tissues. However, future clinical studies and laboratory research are necessary to elucidate the precise cause of psoriasis as well as the underlying genetic and immune signaling pathways driving less common clinical variants and recalcitrant disease.

CT11302082_eTable.jpg

References
  1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157:940-946. doi:10.1001/jamadermatol.2021.2007
  2. Ogdie A, Langan S, Love T, et al. Prevalence and treatment patterns of psoriatic arthritis in the UK. Rheumatology (Oxford). 2013;52:568-575. doi:10.1093/rheumatology/kes324
  3. Mease PJ, Gladman DD, Papp KA, et al. Prevalence of rheumatologistdiagnosed psoriatic arthritis in patients with psoriasis in European/ North American dermatology clinics. J Am Acad Dermatol. 2013;69:729- 735. doi:10.1016/j.jaad.2013.07.023
  4. Langan SM, Seminara NM, Shin DB, et al. Prevalence of metabolic syndrome in patients with psoriasis: a population-based study in the United Kingdom. J Invest Dermatol. 2012;132(3 pt 1):556-562. doi:10.1038/jid.2011.365
  5. Wu S, Li WQ, Han J, et al. Hypercholesterolemia and risk of incident psoriasis and psoriatic arthritis in US women. Arthritis Rheumatol. 2014;66:304-310. doi:10.1002/art.38227
  6. Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and hypertension: a systematic review and meta-analysis of observational studies. J Hypertens. 2013;31:433-442; discussion 442-443. doi:10.1097/HJH.0b013e32835bcce1
  7. Gelfand JM, Dommasch ED, Shin DB, et al. The risk of stroke in patients with psoriasis. J Invest Dermatol. 2009;129:2411-2418. doi:10.1038 /jid.2009.112
  8. Armstrong AW, Lin SW, Chambers CJ, et al. Psoriasis and hypertension severity: results from a case-control study. PLoS One. 2011;6:E18227. doi:10.1371/journal.pone.0018227
  9. Naldi L, Chatenoud L, Linder D, et al. Cigarette smoking, body mass index, and stressful life events as risk factors for psoriasis: results from an Italian case-control study. J Invest Dermatol. 2005;125:61-67. doi:10.1111/j.0022-202X.2005.23681.x
  10. Yang YW, Kang JH, Lin HC. Increased risk of psoriasis following obstructive sleep apnea: a longitudinal population-based study. Sleep Med. 2012;13:285-289. doi:10.1016/j.sleep.2011.07.018
  11. Vaengebjerg S, Skov L, Egeberg A, et al. Prevalence, incidence, and risk of cancer in patients with psoriasis and psoriatic arthritis: a systematic review and meta-analysis. JAMA Dermatol. 2020;156:421-429. doi:10.1001/jamadermatol.2020.0024
  12. Loft N, Skov L, Richardson C, et al. A nationwide population-based cohort study of the incidence of severe and rare infections among adults with psoriasis in Denmark. Br J Dermatol. 2022;187:353-363. doi:10.1111/bjd.21595
  13. Fu Y, Lee CH, Chi CC. Association of psoriasis with inflammatory bowel disease: a systematic review and meta-analysis. JAMA Dermatol. 2018;154:1417-1423. doi:10.1001/jamadermatol.2018.3631
  14. Dowlatshahi EA, Wakkee M, Arends LR, et al. The prevalence and odds of depressive symptoms and clinical depression in psoriasis patients: a systematic review and meta-analysis. J Invest Dermatol. 2014;134:1542-1551. doi:10.1038/jid.2013.508
  15. Kurd SK, Troxel AB, Crits-Christoph P, et al. The risk of depression, anxiety, and suicidality in patients with psoriasis: a populationbased cohort study. Arch Dermatol. 2010;146:891-895. doi:10.1001 /archdermatol.2010.186
  16. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073-1113. doi:10.1016/j.jaad.2018.11.058
  17. Vanderpuye-Orgle J, Zhao Y, Lu J, et al. Evaluating the economic burden of psoriasis in the United States. J Am Acad Dermatol. 2015;72:961-967. doi:10.1016/j.jaad.2015.02.1099
  18. Bos JD, Hagenaars C, Das PK, et al. Predominance of “memory” T cells (CD4+, CDw29+) over “naive” T cells (CD4+, CD45R+) in both normal and diseased human skin. Arch Dermatol Res. 1989;281:24-30. doi:10.1007/BF00424268
  19. Bos JD, Hulsebosch HJ, Krieg SR, et al. Immunocompetent cells in psoriasis. in situ immunophenotyping by monoclonal antibodies. Arch Dermatol Res. 1983;275:181-189. doi:10.1007/BF00510050
  20. Griffiths CE, Powles AV, Leonard JN, et al. Clearance of psoriasis with low dose cyclosporin. Br Med J (Clin Res Ed). 1986;293:731-732. doi:10.1136/bmj.293.6549.731
  21. Ellis CN, Gorsulowsky DC, Hamilton TA, et al. Cyclosporine improves psoriasis in a double-blind study. JAMA. 1986;256:3110-3116.
  22. Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233-240. doi:10.1084/jem.20041257
  23. Ferber IA, Brocke S, Taylor-Edwards C, et al. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol. 1996;156:5-7.
  24. Bharadwaj R, Lusi CF, Mashayekh S, et al. Methotrexate suppresses psoriatic skin inflammation by inhibiting muropeptide transporter SLC46A2 activity. Immunity. 2023;56:998-1012. doi:10.1016/j. immuni.2023.04.001
  25. Jenneck C, Novak N. The safety and efficacy of alefacept in the treatment of chronic plaque psoriasis. Ther Clin Risk Manag. 2007;3:411-420.
  26. Pariser DM, Gordon KB, Papp KA, et al. Clinical efficacy of efalizumab in patients with chronic plaque psoriasis: results from three randomized placebo-controlled phase III trials: part I. J Cutan Med Surg. 2005; 9:303-312. doi:10.1007/s10227-005-0116-1
  27. Mease PJ, Gottlieb AB, van der Heijde D, et al. Efficacy and safety of abatacept, a T-cell modulator, in a randomised, double-blind, placebo-controlled, phase III study in psoriatic arthritis. Ann Rheum Dis. 2017;76:1550-1558. doi:10.1136/annrheumdis-2016-210724
  28. Hawkes JE, Yan BY, Chan TC, et al. Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis. J Immunol. 2018;201:1605-1613. doi:10.4049/jimmunol.1800013
  29. Ogawa K, Okada Y. The current landscape of psoriasis genetics in 2020. J Dermatol Sci. 2020;99:2-8. doi:10.1016/j.jdermsci.2020.05.008
  30. Arakawa A, Siewert K, Stohr J, et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med. 2015;212:2203-2212. doi:10.1084/jem.20151093
  31. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263-271. doi:10.1016/S0140-6736(07)61128-3
  32. Wang CQF, Akalu YT, Suarez-Farinas M, et al. IL-17 and TNF synergistically modulate cytokine expression while suppressing melanogenesis: potential relevance to psoriasis. J Invest Dermatol. 2013;133:2741-2752. doi:10.1038/jid.2013.237
  33. Cheung KL, Jarrett R, Subramaniam S, et la. Psoriatic T cells recognize neolipid antigens generated by mast cell phospholipase delivered by exosomes and presented by CD1a. J Exp Med. 2016;213:2399-2412. doi:10.1084/jem.20160258
  34. Hawkes JE, Gonzalez JA, Krueger JG. Autoimmunity in psoriasis: evidence for specific autoantigens. Curr Dermatol Rep. 2017;6:104-112. doi:10.1007/s13671-017-0177-6
  35. Johansen C, Usher PA, Kjellerup RB, et al. Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. Br J Dermatol. 2009;160:319-324. doi:10.1111/j.1365-2133 .2008.08902.x
  36. Kolbinger F, Loesche C, Valentin MA, et al. beta-Defensin 2 is a responsive biomarker of IL-17A-driven skin pathology in patients with psoriasis. J Allergy Clin Immunol. 2017;139:923-932. doi:10.1016/j .jaci.2016.06.038
  37. Ruddy MJ, Wong GC, Liu XK, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J Biol Chem. 2004;279:2559-2567. doi:10.1074/jbc.M308809200
  38. Shen F, Hu Z, Goswami J, et al. Identification of common transcriptional regulatory elements in interleukin-17 target genes. J Biol Chem. 2006;281:24138-24148. doi:10.1074/jbc.M604597200
  39. Harper EG, Guo C, Rizzo H, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis. J Invest Dermatol. 2009;129:2175-2183. doi:10.1038/jid.2009.65
  40. Chiricozzi A, Guttman-Yassky E, Suarez-Farinas M, et al. Integrative responses to IL-17 and TNF-alpha in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. J Invest Dermatol. 2011;131:677-687. doi:10.1038/jid.2010.340
  41. Homey B, Dieu-Nosjean MC, Wiesenborn A, et al. Up-regulation of macrophage inflammatory protein-3 alpha/CCL20 and CC chemokine receptor 6 in psoriasis. J Immunol. 2000;164:6621-6632. doi:10.4049 /jimmunol.164.12.6621
  42. Stephen-Victor E, Fickenscher H, Bayry J. IL-26: an emerging proinflammatory member of the IL-10 Cytokine family with multifaceted actions in antiviral, antimicrobial, and autoimmune responses. PLoS Pathog. 2016;12:E1005624. doi:10.1371/journal.ppat.1005624
  43. Wolk K, Witte K, Witte E, et al. IL-29 is produced by T(H)17 cells and mediates the cutaneous antiviral competence in psoriasis [published online September 25, 2013]. Sci Transl Med. doi:10.1126 /scitranslmed.3006245
  44. Kasprowicz-Furmanczyk M, Czerwinska J, Placek W, et al. Assessment of the tissue resident memory cells in lesional skin of patients with psoriasis and in healthy skin of healthy volunteers. Int J Environ Res Public Health. 2021;18:11251. doi:10.3390/ijerph182111251
  45. Cheuk S, Schlums H, Gallais Serezal I, et al. CD49a expression defines tissue-resident CD8(+) T cells poised for cytotoxic function in human skin. Immunity. 2017;46:287-300. doi:10.1016/j.immuni.2017.01.009
  46. Sawyer LM, Malottki K, Sabry-Grant C, et al. Assessing the relative efficacy of interleukin-17 and interleukin-23 targeted treatments for moderate-to-severe plaque psoriasis: a systematic review and network meta-analysis of PASI response. PLoS One. 2019;14:E0220868. doi:10.1371/journal.pone.0220868
  47. Coates LC, Kavanaugh A, Mease PJ, et al. Group for research and assessment of psoriasis and psoriatic arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68:1060-1071. doi:10.1002/art.39573
  48. Wang EA, Suzuki E, Maverakis E, et al. Targeting IL-17 in psoriatic arthritis. Eur J Rheumatol. 2017;4:272-277. doi:10.5152/eurjrheum.2017.17037
  49. Armstrong A, Fahrbach K, Leonardi C, et al. Efficacy of bimekizumab and other biologics in moderate to severe plaque psoriasis: a systematic literature review and a network meta-analysis. Dermatol Ther (Heidelb). 2022;12:1777-1792. doi:10.1007/s13555-022-00760-8
  50. van Baarsen LG, Lebre MC, van der Coelen D, et al. Heterogeneous expression pattern of interleukin 17A (IL-17A), IL-17F and their receptors in synovium of rheumatoid arthritis, psoriatic arthritis and osteoarthritis: possible explanation for nonresponse to anti-IL-17 therapy? Arthritis Res Ther. 2014;16:426. doi:10.1186/s13075-014-0426-z
  51. Hot A, Zrioual S, Toh ML, et al. IL-17A- versus IL-17F-induced intracellular signal transduction pathways and modulation by IL-17RA and IL-17RC RNA interference in rheumatoid synoviocytes. Ann Rheum Dis. 2011;70:341-348. doi:10.1136/ard.2010.132233
  52. Adams R, Maroof A, Baker T, et al. Bimekizumab, a novel humanized IgG1 antibody that neutralizes both IL-17A and IL-17F. Front Immunol. 2020;11:1894. doi:10.3389/fimmu.2020.01894
  53. Gordon KB, Foley P, Krueger JG, et al. Bimekizumab efficacy and safety in moderate to severe plaque psoriasis (BE READY): a multicentre, double-blind, placebo-controlled, randomised withdrawal phase 3 trial. Lancet. 2021;397:475-486. doi:10.1016/S0140-6736(21)00126-4
  54. Glatt S, Baeten D, Baker T, et al. Dual IL-17A and IL-17F neutralisation by bimekizumab in psoriatic arthritis: evidence from preclinical experiments and a randomised placebo-controlled clinical trial that IL-17F contributes to human chronic tissue inflammation. Ann Rheum Dis. 2018;77:523-532. doi:10.1136 /annrheumdis-2017-212127
  55. Gordon KB, Langley RG, Warren RB, et al. Bimekizumab safety in patients with moderate to severe plaque psoriasis: pooled results from phase 2 and phase 3 randomized clinical trials. JAMA Dermatol. 2022;158:735-744. doi:10.1001/jamadermatol.2022.1185
  56. Reich K, Warren RB, Lebwohl M, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med. 2021;385:142-152. doi:10.1056/NEJMoa2102383
  57. Reich K, Iversen L, Puig L, et al. Long-term efficacy and safety of brodalumab in moderate-to-severe plaque psoriasis: a post hoc pooled analysis of AMAGINE-2 and -3. J Eur Acad Dermatol Venereol. 2022;36:1275-1283. doi:10.1111/jdv.18068
  58. Papp KA, Blauvelt A, Puig L, et al. Long-term safety and efficacy of risankizumab for the treatment of moderate-to-severe plaque psoriasis: interim analysis of the LIMMitless open-label extension trial up to 5 years of follow-up. J Am Acad Dermatol. 2023;89:1149-1158. doi: 10.1016/j.jaad.2023.07.1024
  59. Glatt S, Jemec GBE, Forman S, et al. Efficacy and safety of bimekizumab in moderate to severe hidradenitis suppurativa: a phase 2, doubleblind, placebo-controlled randomized clinical trial. JAMA Dermatol. 2021;157:1279-1288. doi:10.1001/jamadermatol.2021.2905
  60. Choon SE, Lai NM, Mohammad NA, et al. Clinical profile, morbidity, and outcome of adult-onset generalized pustular psoriasis: analysis of 102 cases seen in a tertiary hospital in Johor, Malaysia. Int J Dermatol. 2014;53:676-684. doi:10.1111/ijd.12070
  61. Zheng M, Jullien D, Eyerich K. The prevalence and disease characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23 (suppl 1):5-12. doi:10.1007/s40257-021-00664-x
  62. Fujita H, Gooderham M, Romiti R. Diagnosis of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23(suppl 1):31-38. doi:10.1007/s40257-021-00652-1
  63. Choon SE, Navarini AA, Pinter A. Clinical course and characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23 (suppl 1):21-29. doi:10.1007/s40257-021-00654-z
  64. Augey F, Renaudier P, Nicolas JF. Generalized pustular psoriasis (Zumbusch): a French epidemiological survey. Eur J Dermatol. 2006;16:669-673.
  65. Ohkawara A, Yasuda H, Kobayashi H, et al. Generalized pustular psoriasis in Japan: two distinct groups formed by differences in symptoms and genetic background. Acta Derm Venereol. 1996;76:68-71. doi:10.2340/00015555766871
  66. Lee JY, Kang S, Park JS, et al. Prevalence of psoriasis in Korea: A population-based epidemiological study using the Korean National Health Insurance database. Ann Dermatol. 2017;29:761-767. doi:10.5021 /ad.2017.29.6.761
  67. Prinz JC, Choon SE, Griffiths CEM, et al. Prevalence, comorbidities and mortality of generalized pustular psoriasis: a literature review. J Eur Acad Dermatol Venereol. 2023;37:256-273. doi:10.1111/jdv.18720
  68. Johnston A, Xing X, Wolterink L, et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol. 2017;140:109-120. doi:10.1016/j.jaci.2016.08.056
  69. Rajan N, Sinclair N, Nakai H, et al. A tale of two sisters: identical IL36RN mutations and discordant phenotypes. Br J Dermatol. 2016;174:417-420. doi:10.1111/bjd.14003
  70. Ly K, Beck KM, Smith MP, et al. Diagnosis and screening of patients with generalized pustular psoriasis. Psoriasis (Auckl). 2019;9:37-42. doi:10.2147/PTT.S181808
  71. Sugiura K. Role of interleukin 36 in generalised pustular psoriasis and beyond. Dermatol Ther (Heidelb). 2022;12:315-328. doi:10.1007 /s13555-021-00677-8
  72. Akiyama M, Takeichi T, McGrath JA, et al. Autoinflammatory keratinization diseases: an emerging concept encompassing various inflammatory keratinization disorders of the skin. J Dermatol Sci. 2018;90:105-111. doi:10.1016/j.jdermsci.2018.01.012
  73. Bachelez H, Choon SE, Marrakchi S, et al. Trial of spesolimab for generalized pustular psoriasis. N Engl J Med. 2021;385:2431-2440. doi:10.1056/NEJMoa2111563
  74. Warren RB, Reich A, Kaszuba A, et al. Imsidolimab, an anti-IL-36 receptor monoclonal antibody for the treatment of generalised pustular psoriasis: results from the phase 2 GALLOP trial. Br J Dermatol. 2023;189:161-169. doi:10.1093/bjd/ljad083
  75. Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. 2017; 18:374-384. doi:10.1038/ni.3691
  76. Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case Control Consortium 2; Strange A, Capon F, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42:985-990. doi:10.1038/ng.694
  77. Enerback C, Sandin C, Lambert S, et al. The psoriasis-protective TYK2 I684S variant impairs IL-12 stimulated pSTAT4 response in skin-homing CD4+ and CD8+ memory T-cells. Sci Rep. 2018;8:7043. doi:10.1038/s41598-018-25282-2
  78. Shimoda K, Kato K, Aoki K, et al. Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function. Immunity. 2000;13:561-571. doi:10.1016/s1074-7613(00)00055-8
  79. Karaghiosoff M, Neubauer H, Lassnig C, et al. Partial impairment of cytokine responses in Tyk2-deficient mice. Immunity. 2000;13:549-560. doi:10.1016/s1074-7613(00)00054-6
  80. Burke JR, Cheng L, Gillooly KM, et al. Autoimmune pathways in mice and humans are blocked by pharmacological stabilization of the TYK2 pseudokinase domain [published online July 24, 2019]. Sci Transl Med. doi:10.1126/scitranslmed.aaw1736
  81. Strober B, Thaci D, Sofen H, et al. Deucravacitinib versus placebo and apremilast in moderate to severe plaque psoriasis: efficacy and safety results from the 52-week, randomized, double-blinded, phase 3 program for evaluation of TYK2 inhibitor psoriasis second trial. J Am Acad Dermatol. 2023;88:40-51. doi:10.1016/j.jaad.2022.08.061
  82. Stein Gold L, Lebwohl M, Menter A, et al. Aerosol foam formulation of fixed combination calcipotriene plus betamethasone dipropionate is highly efficacious in patients with psoriasis vulgaris: pooled data from three randomized controlled studies. J Drugs Dermatol. 2016;15:951-957.
  83. Beranek M, Fiala Z, Kremlacek J, et al. Serum levels of aryl hydrocarbon receptor, cytochromes p450 1a1 and 1b1 in patients with exacerbated psoriasis vulgaris. Folia Biol (Praha). 2018;64:97-102.
  84. Esser C, Rannug A. The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev. 2015;67:259- 279. doi:10.1124/pr.114.009001
  85. Furue M, Uchi H, Mitoma C, et al. Antioxidants for healthy skin: the emerging role of aryl hydrocarbon receptors and nuclear factorerythroid 2-related factor-2. Nutrients. 2017;9:223. doi:10.3390/nu9030223
  86. Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008;371:1675-1684. doi:10.1016/S0140-6736(08)60726-6
  87. Sutter CH, Olesen KM, Bhuju J, et al. AHR regulates metabolic reprogramming to promote SIRT1-dependent keratinocyte differentiation. J Invest Dermatol. 2019;139:818-826. doi:10.1016/j.jid.2018.10.019
  88. Haas K, Weighardt H, Deenen R, et al. Aryl hydrocarbon receptor in keratinocytes is essential for murine skin barrier integrity. J Invest Dermatol. 2016;136:2260-2269. doi:10.1016/j.jid.2016.06.627
  89. Di Meglio P, Duarte JH, Ahlfors H, et al. Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity. 2014;40:989-1001. doi:10.1016/j.immuni.2014.04.019
  90. Kim HO, Kim JH, Chung BY, et al. Increased expression of the aryl hydrocarbon receptor in patients with chronic inflammatory skin diseases. Exp Dermatol. 2014;23:278-281. doi:10.1111/exd.12350
  91. van den Bogaard EH, Bergboer JG, Vonk-Bergers M, et al. Coal tar induces AHR-dependent skin barrier repair in atopic dermatitis. J Clin Invest. 2013;123:917-927. doi:10.1172/JCI65642
  92. Smith SH, Jayawickreme C, Rickard DJ, et al. Tapinarof is a natural AHR agonist that resolves skin inflammation in mice and humans. J Invest Dermatol. 2017;137:2110-2119. doi:10.1016/j.jid.2017.05.004
  93. Strober B, Stein Gold L, Bissonnette R, et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J Am Acad Dermatol. 2022;87:800-806. doi:10.1016/j.jaad.2022.06.1171
  94. Mooney N, Teague JE, Gehad AE, et al. Tapinarof inhibits the formation, cytokine production, and persistence of resident memory T cells in vitro. SKIN J Cutan Med. 2023;7:S194. doi:10.25251/skin.7.supp.194
  95. Schafer PH, Truzzi F, Parton A, et al. Phosphodiesterase 4 in inflammatory diseases: effects of apremilast in psoriatic blood and in dermal myofibroblasts through the PDE4/CD271 complex. Cell Signal. 2016;28:753-763. doi:10.1016/j.cellsig.2016.01.007
  96. Li H, Zuo J, Tang W. Phosphodiesterase-4 inhibitors for the treatment of inflammatory diseases. Front Pharmacol. 2018;9:1048. doi:10.3389/ fphar.2018.01048
  97. Schafer PH, Parton A, Gandhi AK, et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br J Pharmacol. 2010;159:842-855. doi:10.1111/j.1476-5381.2009.00559.x
  98. Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol. 2015;73:37-49. doi:10.1016/j .jaad.2015.03.049
  99. Papp KA, Gooderham M, Droege M, et al. Roflumilast cream improves signs and symptoms of plaque psoriasis: results from a phase 1/2a randomized, controlled study. J Drugs Dermatol. 2020;19:734-740. doi:10.36849/JDD.2020.5370
  100. Lebwohl MG, Kircik LH, Moore AY, et al. Effect of roflumilast cream vs vehicle cream on chronic plaque psoriasis: the DERMIS-1 and DERMIS-2 randomized clinical trials. JAMA. 2022;328:1073-1084. doi:10.1001/jama.2022.15632
Article PDF
Author and Disclosure Information

Dr. Nong is from the Department of Internal Medicine, SUNY Downstate Medical Center, Brooklyn, New York. Dr. Nong also is from and Dr. Hawkes is from Integrative Skin Science and Research, Pacific Skin Institute, Sacramento, California. Dr. Han is from the Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York.

Dr. Nong reports no conflict of interest. Dr. Han is or has been an investigator, consultant/advisor, or speaker for AbbVie, Amgen, Arcutis, Bausch Health, Boehringer Ingelheim, Bristol Myers Squibb, Dermavant, DermTech, Eli Lilly and Company, EPI Health, Janssen Pharmaceuticals, LEO Pharma, Novartis, Ortho Dermatologics, Pfizer Inc, Regeneron Pharmaceuticals, Sanofi Genzyme, Sun Pharmaceutical Industries Ltd, and UCB. He also has received research grants from Athenex, Bausch Health, Bond Avillion, Eli Lilly and Company, Janssen Pharmaceuticals, MC2 Therapeutics, Novartis, PellePharm, and Pfizer Inc. Dr. Hawkes is a consultant/advisor for AbbVie, Arcutis Biotherapeutics, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly and Company, Janssen Pharmaceuticals, LEO Pharma, Novartis, Pfizer, Regeneron Pharmaceuticals, Sanofi, Sun Pharmaceutical Industries Ltd, and UCB. He also is a speaker for Boehringer Ingelheim, Bristol Myers Squibb, Regeneron Pharmaceuticals, Sanofi, and UCB.

The eTable is in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Jason E. Hawkes, MD, MS, Integrative Skin Science and Research, Pacific Skin Institute, 1495 River Park Dr, Sacramento, CA 95815 (hawkes3@gmail.com).

Issue
Cutis - 113(2)
Publications
Topics
Page Number
82-91,E3
Sections
Author and Disclosure Information

Dr. Nong is from the Department of Internal Medicine, SUNY Downstate Medical Center, Brooklyn, New York. Dr. Nong also is from and Dr. Hawkes is from Integrative Skin Science and Research, Pacific Skin Institute, Sacramento, California. Dr. Han is from the Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York.

Dr. Nong reports no conflict of interest. Dr. Han is or has been an investigator, consultant/advisor, or speaker for AbbVie, Amgen, Arcutis, Bausch Health, Boehringer Ingelheim, Bristol Myers Squibb, Dermavant, DermTech, Eli Lilly and Company, EPI Health, Janssen Pharmaceuticals, LEO Pharma, Novartis, Ortho Dermatologics, Pfizer Inc, Regeneron Pharmaceuticals, Sanofi Genzyme, Sun Pharmaceutical Industries Ltd, and UCB. He also has received research grants from Athenex, Bausch Health, Bond Avillion, Eli Lilly and Company, Janssen Pharmaceuticals, MC2 Therapeutics, Novartis, PellePharm, and Pfizer Inc. Dr. Hawkes is a consultant/advisor for AbbVie, Arcutis Biotherapeutics, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly and Company, Janssen Pharmaceuticals, LEO Pharma, Novartis, Pfizer, Regeneron Pharmaceuticals, Sanofi, Sun Pharmaceutical Industries Ltd, and UCB. He also is a speaker for Boehringer Ingelheim, Bristol Myers Squibb, Regeneron Pharmaceuticals, Sanofi, and UCB.

The eTable is in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Jason E. Hawkes, MD, MS, Integrative Skin Science and Research, Pacific Skin Institute, 1495 River Park Dr, Sacramento, CA 95815 (hawkes3@gmail.com).

Author and Disclosure Information

Dr. Nong is from the Department of Internal Medicine, SUNY Downstate Medical Center, Brooklyn, New York. Dr. Nong also is from and Dr. Hawkes is from Integrative Skin Science and Research, Pacific Skin Institute, Sacramento, California. Dr. Han is from the Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York.

Dr. Nong reports no conflict of interest. Dr. Han is or has been an investigator, consultant/advisor, or speaker for AbbVie, Amgen, Arcutis, Bausch Health, Boehringer Ingelheim, Bristol Myers Squibb, Dermavant, DermTech, Eli Lilly and Company, EPI Health, Janssen Pharmaceuticals, LEO Pharma, Novartis, Ortho Dermatologics, Pfizer Inc, Regeneron Pharmaceuticals, Sanofi Genzyme, Sun Pharmaceutical Industries Ltd, and UCB. He also has received research grants from Athenex, Bausch Health, Bond Avillion, Eli Lilly and Company, Janssen Pharmaceuticals, MC2 Therapeutics, Novartis, PellePharm, and Pfizer Inc. Dr. Hawkes is a consultant/advisor for AbbVie, Arcutis Biotherapeutics, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly and Company, Janssen Pharmaceuticals, LEO Pharma, Novartis, Pfizer, Regeneron Pharmaceuticals, Sanofi, Sun Pharmaceutical Industries Ltd, and UCB. He also is a speaker for Boehringer Ingelheim, Bristol Myers Squibb, Regeneron Pharmaceuticals, Sanofi, and UCB.

The eTable is in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Jason E. Hawkes, MD, MS, Integrative Skin Science and Research, Pacific Skin Institute, 1495 River Park Dr, Sacramento, CA 95815 (hawkes3@gmail.com).

Article PDF
Article PDF

Psoriasis is a chronic inflammatory disease that affects approximately 3% of the US population.1 Plaque psoriasis comprises 80% to 90% of cases, while pustular, erythrodermic, guttate, inverse, and palmoplantar disease are less common variants (Figure 1). Psoriatic skin manifestations range from localized to widespread or generalized disease with recurrent flares. Body surface area or psoriasis area and severity index (PASI) measurements primarily focus on skin manifestations and are important for evaluating disease activity and response to treatment, but they have inherent limitations: they do not capture extracutaneous disease activity, systemic inflammation, comorbid conditions, quality of life impact, or the economic burden of psoriasis.

CT11302082_ABCD.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Characteristic%20plaque%20psoriasis%20of%20the%20trunk.%20C%2C%20Inverse%20psoriasis%20involving%20the%20inframammary%20folds.%20D%2C%20Guttate%20psoriasis%20in%20an%20adult%20following%20streptococcal%20infection.%3C%2Fp%3E

A common manifestation of psoriasis is psoriatic arthritis (PsA), which can involve the nails, joints, ligaments, or tendons in 30% to 41% of affected individuals (Figure 2).2,3 A growing number of psoriasis-associated comorbidities also have been reported including metabolic syndrome4; hyperlipidemia5; cardiovascular disease6; stroke7; hypertension8; obesity9; sleep disorders10; malignancy11; infections12; inflammatory bowel disease13; and mental health disorders such as depression,14 anxiety,15 and suicidal ideation.15 Psoriatic disease also interferes with daily life activities and a patient’s overall quality of life, including interpersonal relationships, intimacy, employment, and work productivity.16 Finally, the total estimated cost of psoriasis-related health care is more than $35 billion annually,17 representing a substantial economic burden to our health care system and individual patients.

Nong_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Clinical%20manifestation%20of%20psoriatic%20arthritis%20involving%20the%20metacarpal%20joints%20of%20the%20hands.%3C%2Fp%3E

The overall burden of psoriatic disease has declined markedly in the last 2 decades due to revolutionary advances in our understanding of the immunopathogenesis of psoriasis and the subsequent development of improved therapies that predominantly interrupt IL-23/IL-17 cytokine signaling; however, critical knowledge and treatment gaps persist, underscoring the importance of ongoing clinical and research efforts in psoriatic disease. We review the working immune model of psoriasis, summarize related immune discoveries, and highlight recent therapeutic innovations that are shaping psoriatic disease management.

Current Immune Model of Psoriatic Disease

Psoriasis is an autoinflammatory T cell–mediated disease with negligible contributions from the humoral immune response. Early clinical observations reported increased inflammatory infiltrates in psoriatic skin lesions primarily consisting of both CD4+ and CD8+ T-cell populations.18,19 Additionally, patients treated with broad-acting, systemic immunosuppressive medications (eg, cyclosporine, oral corticosteroids) experienced improvement of psoriatic lesions and normalization of the immune infiltrates observed in skin biopsy specimens.20,21 These early clinical findings led to more sophisticated experimentation in xenotransplant models of psoriasis,22,23 which explored the clinical efficacy of several less immunosuppressive (eg, methotrexate, anti–tumor necrosis factor [TNF] biologics)24 or T cell–specific agents (eg, alefacept, abatacept, efalizumab).25-27 The results of these translational studies provided indisputable evidence for the role of the dysregulated immune response as the primary pathogenic process driving plaque formation; they also led to a paradigm shift in how the immunopathogenesis of psoriatic disease was viewed and paved the way for the identification and targeting of other specific proinflammatory signals produced by activated dendritic cell (DC) and T-lymphocyte populations. Among the psoriasis-associated cytokines subsequently identified and studied, elevated IL-23 and IL-17 cytokine levels in psoriatic skin were most closely associated with disease activity, and rapid normalization of IL-23/IL-17 signaling in response to effective oral or injectable antipsoriatic treatments was the hallmark of skin clearance.28 The predominant role of IL-23/IL-17 signaling in the development and maintenance of psoriatic disease is the central feature of all working immune models for this disease (Figure 3).

Nong_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Working%20immune%20model%20of%20psoriasis.%20Early%20immune%20events%20include%20activation%20of%20dendritic%20cells%20(DCs)%20and%20IL-17%E2%80%93producing%20T%20cells%20(T17)%20in%20the%20prepsoriatic%20(or%20normal-appearing)%20skin%20of%20individuals%20who%20are%20genetically%20susceptible%20and%2For%20have%20exposures%20to%20known%20psoriasis%20triggers.%20Activation%20of%20DC%20and%20T17%20populations%20in%20the%20skin%20results%20in%20increased%20production%20of%20tumor%20necrosis%20factor%20(TNF)%2C%20IL-23%2C%20and%20IL-17%20cytokines%20(namely%20IL-17A%20and%20IL-17F)%2C%20which%20work%20synergistically%20with%20other%20immune%20signals%20(IL-12%2C%20IL-22%2C%20IL-36%2C%20TNF%2C%20interferon%20%5BIFN%5D)%20to%20drive%20keratinocyte%20(KC)%20hyperproliferation.%20In%20response%20to%20upregulated%20IL-17%20signaling%2C%20substantial%20increases%20in%20keratinocyte-derived%20proteins%20(antimicrobial%20peptides%2C%20IL-19%2C%20IL-36%2C%20IL-17C)%20and%20chemotactic%20factors%20(chemokine%20%5BC-C%20motif%5D%20ligand%2020%20%5BCCL20%5D%2C%20chemokine%20%5BC-C%20motif%5D%20ligand%201%2F2%2F3%2F5%2F8%20%5BCXCL1%2F2%2F3%2F5%2F8%5D%5Bor%20IL-8%5D)%20facilitate%20further%20activation%20and%20recruitment%20of%20T17%20and%20helper%20T%20cell%20(TH1)%20lymphocytes%2C%20DCs%2C%20macrophages%2C%20and%20polymorphonuclear%20neutrophils%20(PMNs)%20into%20the%20skin.%20The%20resultant%20inflammatory%20circuit%20creates%20a%20self-amplifying%20or%20feed-forward%20immune%20response%20in%20the%20skin%20that%20leads%20to%20the%20hallmark%20clinical%20features%20of%20psoriasis%20and%20sustains%20the%20mature%20psoriatic%20plaque.%3C%2Fp%3E

Psoriasis-Associated Genetic and Environmental Risk Factors

The exact sequence of events that lead to the initiation and formation of plaque psoriasis in susceptible individuals is still poorly understood; however, several important risk factors and key immune events have been identified. First, decades of genetic research have reported more than 80 known psoriasis-associated susceptibility loci,29 which explains approximately 50% of psoriasis heritability. The major genetic determinant of psoriasis, HLA-C*06:02 (formerly HLA-Cw6), resides in the major histocompatibility complex class I region on chromosome 6p21.3 (psoriasis susceptibility gene 1, PSORS1) and is most strongly associated with psoriatic disease.30 Less common psoriasis-associated susceptibility genes also are known to directly or indirectly impact innate and adaptive immune functions that contribute to the pathogenesis of psoriasis.

Second, several nongenetic environmental risk factors for psoriasis have been reported across diverse patient populations, including skin trauma/injury, infections, alcohol/tobacco use, obesity, medication exposure (eg, lithium, antimalarials, beta-blockers), and stress.31 These genetic and/or environmental risk factors can trigger the onset of psoriatic disease at any stage of life, though most patients develop disease in early adulthood or later (age range, 50–60 years). Some patients never develop psoriasis despite exposure to environmental risk factors and/or a genetic makeup that is similar to affected first-degree relatives, which requires further study.

Prepsoriatic Skin and Initiation of Plaque Development

In response to environmental stimuli and/or other triggers of the immune system, DC and resident IL-17–producing T-cell (T17) populations become activated in predisposed individuals. Dendritic cell activation leads to the upregulation and increase of several proinflammatory cytokines, including TNF, interferon (IFN) α, IFN-γ, IL-12, and IL-23. Tumor necrosis factor and IL-23 play a vital role in psoriasis by helping to regulate the polarization and expansion of T22 and T17 cells in the skin, whereas IL-12 promotes a corresponding type 1 inflammatory response.32 Increased IL-17 and IL-22 result in alteration of the terminal differentiation and proliferative potential of epidermal keratinocytes, leading to the early clinical hallmarks of psoriatic plaques. The potential contribution of overexpressed psoriasis-related autoantigens, such as LL-37/cathelicidin, ADAMTSL5, and PLA2G4D,33 in the initiation of psoriatic plaques has been suggested but is poorly characterized.34 Whether these specific autoantigens or others presented by HLA-C variants found on antigen-presenting cells are required for the breakdown of immune tolerance and psoriatic disease initiation is highly relevant but requires further investigation and validation.

 

 

Feed-Forward Inflammation, Mature Psoriatic Plaques, and Resident Memory T Cells

In response to the upstream production of IL-23 by dermal DCs, high levels of IL-17 cytokines can be found in mature psoriatic plaques. The IL-17 family consists of 6 dimeric cytokines (IL-17A through IL-17F) that provide innate cutaneous protection against bacterial, viral, and fungal infectious agents, such as Candida albicans. Unlike other IL-17 isoforms, IL-17A and IL-17F share the same receptor complex and have the highest structural homology of any pair (approximately 50% similar).35 The relative expression of IL-17F is higher than IL-17A in psoriasis,36 though IL-17A has been considered as the predominant IL-17 cytokine found in psoriatic skin lesions due to its higher potency.

Binding of IL-17A/F with the IL-17 receptor (IL-17R) on keratinocytes contributes to the development of psoriatic plaques by inducing epidermal hyperplasia via activation of CCAAT/enhancer-binding proteins β and δ, nuclear factor κB, and signal transducer and activator of transcription 1 gene (STAT1).37,38 This also increases the expression of other keratinocyte-derived proteins (eg, human β-defensins, S-100 proteins, LL-37, other antimicrobial peptides, IL-19, IL-36, IL-17C) that act as reinforcing proinflammatory signals or chemotactic factors (eg, chemokine [C-C motif] ligand 20 [CCL20], chemokine [C-C motif] ligand 1/2/3/5 [CXCL1/2/3/5], CXCL8, IL-8) that facilitate the recruitment of additional immune cells to the skin including polymorphonuclear neutrophils (PMNs), macrophages, and DCs.39-41 Routine immunohistochemical staining for these keratinocyte-derived proteins reveals a striking epidermal gene expression gradient wherein levels of IL-17–induced proteins are most highly expressed in the uppermost layers of keratinocytes and facilitate the recruitment of immune cells into the epidermis. Activated T17 cells also stimulate the production of keratinocyte-derived chemokines (eg, CXCL9/10/11), which recruit type 1 inflammatory T-cell populations into developing psoriatic plaques.42,43 Finally, TNF, IL-36, and IL-17C cytokines act synergistically with IL-17A/F to amplify the proinflammatory effects of IL-17 signaling and further stimulate their production from T17 cell populations.40 This inflammatory circuit in the skin creates and supports a self-amplifying or positive feedback loop between the skin and immune system that commonly is referred to as feed-forward inflammation (Figure 3).34 The feed-forward inflammatory loop in psoriasis—predominantly driven by increased IL-23/IL-17 signaling—best characterizes the mature psoriatic plaque.

Several findings suggest that the influx of persistent, long-lived resident memory T cells (Trms) may contribute to the mature psoriatic plaque. It is believed that CD8+CD103+CD49a Trm cell populations may be responsible for the sharply demarcated borders of untreated psoriasis plaques or their recurrence at specific body sites such as the scalp, buttocks, extremity extensor surfaces, umbilicus, or acral skin following specific stimuli or trauma (Koebner phenomenon or isomorphic response).44,45 It is not known if repeated stimuli or trauma induce disease formation via the activation of Trm cell populations; further study in large patient cohorts is needed, but this remains an intriguing area of study for durable treatment responses and potential cures for psoriasis.

Recent Discoveries in Psoriatic Disease

Remarkable treatment outcomes for psoriasis have been achieved with multiple selective IL-17 and IL-23 inhibitors (eTable). As demonstrated in several pivotal phase 3 clinical trials for members of these classes of medications, the majority of treated psoriasis patients achieved PASI90 clearance.46 Due to their more favorable dosing schedule (ie, fewer injections) and ability to induce a durable remissionlike treatment response, IL-23 inhibitors have become the preferred treatment class for cutaneous disease, while IL-17 inhibitors may be preferred when treating patients with both plaque psoriasis and PsA.47,48 Nevertheless, the complexity of this disease is punctuated by treated patients who do not adequately respond to selective IL-23/IL-17 blockade.49 Recent and emerging treatments may shed light on these recalcitrant cases and will add to the rapidly growing arsenal of available psoriasis therapies.

The Role of IL-17F in Psoriasis and Other Inflammatory Skin Diseases

Dysregulation of IL-17A and IL-17F is associated with several chronic inflammatory conditions, such as psoriasis and PsA.35,50 Both cytokines, either as homodimers or heterodimers, can selectively bind to the heterodimeric IL-17R formed by the IL-17RA and IL-17RC subunits.35 IL-17F and IL-17C also can synergize with TNF and other cytokines to promote and support the self-sustaining inflammatory circuits in mature psoriatic plaques, though their inflammatory effects in the skin are more limited than IL-17A.51,52 Therefore, incomplete blockade of IL-17 signaling (ie, unopposed IL-17F and IL-17C) represents a potential mechanism to explain the persistence of psoriasis in patients treated with selective IL-17A inhibitors. This hypothesis is supported by reports of psoriasis patients who have inadequate clinical responses to selective IL-17A inhibition but subsequently improve with IL-17R blockade, which results in disruption of IL-17A as well as IL-17C/E/F cytokine signaling. This formed the basis for further study into the specific role of IL-17F in psoriatic disease and any potential therapeutic benefits associated with its inhibition.

Recently approved in the European Union, Canada, Australia, Japan, the United Kingdom, and the United States for moderate to severe psoriasis, bimekizumab is a novel humanized IgG antibody that selectively inhibits both IL-17A and IL-17F cytokines.53 Specifically, bimekizumab simultaneously prevents binding of IL-17A/A, IL-17A/F, and IL-17F/F dimers with the IL-17R. Compared to other IL-17 and IL-23 biologic therapies, bimekizumab (320 mg) achieved relatively higher response rates for PASI75, PASI90, and PASI100.49 Neutralization of IL-17A and IL-17F by bimekizumab also resulted in more complete suppression of cytokine responses and PMN chemotaxis than either cytokine alone in treated PsA patients,54 which is notable because of the incremental benefits of recent IL-23 and IL-17 inhibitors on inflammatory arthritis symptoms in contrast to the substantial improvements observed for cutaneous disease with those same agents.

The primary disadvantage of bimekizumab and its more complete blockade of the IL-17 signaling pathway is that treated patients have a substantially increased risk for oral candidiasis (>10%).55 However, the precise link between candidiasis and IL-17 blockade is not yet fully understood because other targeted agents that also broadly suppress IL-17 signaling (ie, IL-17R, IL-23 inhibitors) are associated with much lower rates of candidiasis.56-58 Bimekizumab also is being investigated as a novel therapy for hidradenitis suppurativa and will provide important reference information regarding the role for bispecific biologic agents in the treatment of chronic inflammatory skin diseases.59

 

 

IL-36 Signaling and Generalized Pustular Psoriasis

Recent genetic and clinical studies have expanded our understanding of the role of IL-36 signaling in the immunopathogenesis of pustular psoriasis variants. Generalized pustular psoriasis (GPP) is a rare distinct psoriasis subtype characterized by the recurrent development of widespread erythema, superficial sterile pustules, and desquamation. Systemic symptoms such as fever, malaise, itching, and skin pain accompany acute GPP flares.60 Generalized pustular psoriasis is more common in female patients (in contrast with plaque psoriasis), and acute flares may be caused by multiple stimuli including infections, hypocalcemia, initiation or discontinuation of medications (eg, oral corticosteroids), pregnancy, or stress.61,62 Flares of GPP often require emergency or in-patient care, as untreated symptoms increase the risk for severe health complications such as secondary infections, sepsis, or multisystem organ failure.63 The prevalence of GPP is estimated to be approximately 1 in 10,000 individuals in the United States,64-67 with mortality rates ranging from 0 to 3.3 deaths per 100 patient-years.67

In contrast to plaque psoriasis, aberrant IL-36 signaling is the predominant driver of GPP. IL-36 is a member of the IL-1 cytokine family that includes three IL-36 agonists (IL-36α, IL-36β, IL-36γ) and 1 endogenous antagonist (IL-36Ra, encoded by IL36RN).68 The immunopathogenesis of GPP involves dysregulation of the IL-36–chemokine–PMN axis, resulting in unopposed IL-36 signaling and the subsequent recruitment and influx of PMNs into the epidermis. IL36RN mutations are strongly associated with GPP and result in impaired function of the IL-36Ra protein, leading to unopposed IL-36 signaling.69 However, approximately two-thirds of GPP patients lack identifiable gene mutations, suggesting other immune mechanisms or triggers causing upregulated IL-36 signaling.70 In response to these triggers, increased IL-36 cytokines released by keratinocytes bind to the IL-36R, resulting in substantial keratinocyte hyperproliferation, increased IL-36 levels, and the expression of hundreds of additional inflammatory signals (eg, IL-17C, antimicrobial peptides, TNF, IL-6).71 Increased IL-36 levels also drive the production of PMN chemotactic proteins (eg, CXCL1/2/3/5/6/8 and CXCR1/2) and act synergistically with IL-17 cytokines to create an autoamplifying circuit that is analogous to the feed-forward inflammatory loop in plaque psoriasis.72 Biopsies of involved GPP skin reveal increased expression of IL-36 in the uppermost layers of the epidermis, which creates a gene expression gradient that acts as a strong attractant for PMNs and forms the basis for the hallmark pustular lesions observed in GPP patients.

Until recently, treatment strategies for GPP involved the off-label use of topical, oral, or biologic therapies approved for plaque psoriasis, which often was associated with variable or incomplete disease control. In September 2022, the US Food and Drug Administration (FDA) approved intravenous spesolimab as a first-in-class humanized monoclonal IgG1 antibody for the treatment of GPP flares in adults. Spesolimab binds to IL-36R and prevents its activation by its endogenous agonists. A phase 2, randomized, 12-week clinical trial (Effisayil-1) evaluated the efficacy and safety of a single 900-mg intravenous dose of spesolimab followed by an optional second dose 1 week later for inadequate treatment responses in 53 enrolled GPP patients (2:1 treatment to placebo randomization).73 Remarkably, more than half (19/35 [54%]) of GPP patients experienced complete resolution of pustules (GPP physician global assessment subscore of 0 [range, 0–4]) and showed sustained efficacy out to week 12 after just 1 or 2 doses of spesolimab. Overall, the safety profile of spesolimab was good; asthenia, fatigue, nausea, vomiting, headache, pruritus, infusion-related reaction and symptoms, and mild infections (eg, urinary tract infection) were the most common adverse events reported.73

Imsidolimab, a high-affinity humanized IgG4 monoclonal antibody that binds and blocks activation of IL-36R, also has completed phase 2 testing,74 with phase 3 study results expected in early 2024. The rapid onset of action and overall safety of imsidolimab was in line with and similar to spesolimab. Future approval of imsidolimab would add to the limited treatment options available for GPP and has the additional convenience of being administered to patients subcutaneously. Overall, the development of selective IL-36R inhibitors offers a much-needed therapeutic option for GPP and illustrates the importance of translational research.

Role of Tyrosine Kinase in Psoriatic Disease

The Janus kinase (JAK) enzyme family consists of 4 enzymes—tyrosine kinase 2 (TYK2), JAK1, JAK2, and JAK3—that function as intracellular transduction signals that mediate the biologic response of most extracellular cytokines and growth factors.75 Critical psoriasis-related cytokines are dependent on intact JAK-STAT signaling, including IL-23, IL-12, and type I IFNs. In 2010, a genome-wide association identified TYK2 as a psoriasis susceptibility locus,76 and loss-of-function TYK2 mutations confer a reduced risk for psoriasis.77 Unlike other JAK isoforms, TYK2 mediates biologic functions that are highly restricted to the immune responses associated with IL-23, IL-12, and type I IFN signaling.78,79 For these reasons, blockade of TYK2 signaling is an attractive therapeutic target for the potential treatment of psoriatic disease.

In September 2022, the FDA approved deucravacitinib as a first-in-class, oral, selective TYK2 inhibitor for the treatment of adult patients with moderate to severe plaque psoriasis. It was the first FDA approval of an oral small-molecule treatment for plaque psoriasis in nearly a decade. Deucravacitinib inhibits TYK2 signaling via selective binding of its unique regulatory domain, resulting in a conformational (allosteric) change that interferes with its active domain.80 This novel mechanism of action limits the unwanted blockade of other broad biologic processes mediated by JAK1/2/3. Of note, the FDA did not issue any boxed warnings for deucravacitinib as it did for other FDA-approved JAK inhibitors.

In a head-to-head, 52-week, double-blind, prospective, randomized, phase 3 study, deucravacitinib showed clear superiority over apremilast for PASI75 at week 16 (53.0% [271/511] vs 39.8% [101/254]) and week 24 (58.7% [296/504] vs 37.8% [96/254]).81 Clinical responses were sustained through week 52 and showed efficacy for difficult-to-treat areas such as the scalp, acral sites, and nails. Other advantages of deucravacitinib include once-daily dosing with no need for dose titration or adjustments for renal insufficiency as well as the absence of statistically significant differences in gastrointestinal tract symptoms compared to placebo. The most common adverse effects included nasopharyngitis, upper respiratory tract infections, headache, diarrhea, and herpes infections.81 The potential benefit of deucravacitinib for PsA and psoriasis comorbidities remains to be seen, but it is promising due to its simultaneous disruption of multiple psoriasis-related cytokine networks. Several other TYK2 inhibitors are being developed for psoriatic disease and related inflammatory conditions, underscoring the promise of targeting this intracellular pathway.

 

 

Aryl Hydrocarbon Receptor Agonism

Topical steroids are the mainstay treatment option for localized or limited plaque psoriasis due to their potent immunosuppressive effect on the skin and relatively low cost. Combined with vitamin D analogs, topical steroids result in marked improvements in disease severity and improved tolerability.82 However, chronic use of topical steroids is limited by the need for twice-daily application, resulting in poor treatment compliance; loss of efficacy over time; risk for steroid-induced skin atrophy on special body sites; and patient concerns of potential systemic effects. The discovery of novel drug targets amenable to topical inhibition is needed.

Dysregulated aryl hydrocarbon receptor (AHR) levels have been reported in atopic dermatitis and psoriasis.83 Aryl hydrocarbon receptors are ubiquitously expressed in many cell types and play an integral role in immune homeostasis within the skin, skin barrier function, protection against oxidative stressors, and regulation of proliferating melanocytes and keratinocytes.84,85 They are widely expressed in multiple immune cell types (eg, antigen-presenting cells, T lymphocytes, fibroblasts) and modulate the differentiation of T17 and T22 cells as well as their balance with regulatory T-cell populations.86 In keratinocytes, AHR helps to regulate terminal differentiation, enhance skin barrier integrity via AHR-dependent filaggrin (FLG) expression, and prevent transepidermal water loss.87,88 The mechanisms by which AHR ligands lead to the upregulation or downregulation of specific genes is intricate and highly context dependent, such as the specific ligand and cell type involved. In preclinical studies, AHR-deficient mice develop psoriasiform skin inflammation, increased IL-17 and IL-22 expression, and abnormal skin barrier function.89 Keratinocytes treated with AHR ligands in vitro modulated psoriasis-associated inflammatory cytokines, such as IL-6, IL-8, and type I and II IFNs.89,90 The use of coal tar, one of the earliest historical treatments for psoriasis, is thought to activate AHRs in the skin via organic compound mixtures containing polyaromatic hydrocarbons that help normalize the proinflammatory environment in psoriatic skin.91

In June 2022, the FDA approved tapinarof as a first-in-class, topical, nonsteroidal AHR agonist for the treatment of plaque psoriasis in adults. Although the exact mechanism of action for tapinarof has not been fully elucidated, early studies suggest that its primary function is the activation of AHR, leading to reduced T-cell expansion and T17 cell differentiation. In the imiquimod mouse model, cytokine expression of IL-17A, IL-17F, IL-19, IL-22, IL-23A, and IL-lβ in psoriasiform skin lesions were downregulated following tapinarof treatment.92 In humans, tapinarof treatment is associated with a remittive effect, in which the average time for tapinarof-treated psoriasis lesions to remain clear was approximately 4 months.93 Preliminary research investigating the mechanism by which tapinarof induces this remittive effect is ongoing and may involve the reduced activation and influx of T17 and Trm populations into the skin.94 However, these preclinical studies were performed on healthy dermatome-derived skin tissue cultured in T17-skewing conditions and needs to be replicated in larger samples sizes using human-derived psoriatic tissue. Alternatively, a strong inhibitory effect on IL-23 cytokine signaling may, in part, explain the remittive effect of tapinarof, as an analogous response is observed in patients who start and discontinue treatment with selective IL-23 antagonists. Regardless, the once-daily dosing of tapinarof and sustained treatment response is appealing to psoriasis patients. Tapinarof generally is well tolerated with mild folliculitis (>20% of patients) and contact dermatitis (5% of patients) reported as the most common skin-related adverse events.

New Roles for Phosphodiesterase 4 Inhibition

Phosphodiesterases (PDEs) are enzymes that hydrolyze cyclic nucleotides (eg, cyclic adenosine monophosphate) to regulate intracellular secondary messengers involved in the inflammatory response. One of several enzymes in the PDE family, PDE4, has been shown to have greater activity in psoriatic skin compared to healthy skin.95 Phosphodiesterase inhibitors decrease the degradation of cyclic adenosine monophosphate, which triggers protein kinase A to downregulate proinflammatory (eg, TNF-α, IL-6, IL-17, IL-12, IL-23) cytokines and increased expression of anti-inflammatory signals such as IL-10.96,97 Apremilast, the first oral PDE4 inhibitor approved by the FDA for psoriasis, offered a safe alternative to traditional oral immunosuppressive agents that had extensive risks and potential end-organ adverse effects. Unfortunately, apremilast demonstrated modest efficacy for psoriatic disease (better efficacy in the skin vs joint manifestations) and was supplanted easily by next-generation targeted biologic agents that were more efficacious and lacked the troublesome gastrointestinal tract adverse effects of PDE4 inhibition.98

Crisaborole became the first topical PDE4 inhibitor approved in the United States in December 2016 for twice-daily treatment of atopic dermatitis. Although phase 2 trial results were reported in psoriasis, this indication was never pursued, presumably due to similar improvements in primary outcome measures at week 12, compared to placebo (ClinicalTrials.gov Identifier NCT01300052).

In July 2022, the first topical PDE4 inhibitor indicated for plaque psoriasis was approved by the FDA—­roflumilast cream 0.3% for once-daily use in individuals 12 years and older. Roflumilast was found to be clinically efficacious as early as 2 weeks after its use in an early-phase clinical trial.99 In 2 phase 3 clinical trials (DERMIS-1 and DERMIS-2), roflumilast significantly increased the proportion of patients achieving PASI75 at week 8 compared to vehicle (39%–41.6% vs 5.3%–7.6%, respectively)(P<.001).100 Overall, this nonsteroidal topical therapy was found to be well tolerated, with infrequent reports of application site pain or irritation as adverse events. Similar to tapinarof, patients can apply roflumilast on all body surface areas including the face, external genitalia, and other intertriginous areas.100 Importantly, the broad immune impact of PDE4 inhibition suggests that topical roflumilast likely will be an effective treatment for several additional inflammatory conditions, including seborrheic dermatitis and atopic dermatitis, which would expand the clinical utility of this specific medication.

Conclusion

In the last 2 decades, we have witnessed a translational revolution in our understanding of the underlying genetics and immunology of psoriatic disease. Psoriasis is widely considered one of the best-managed inflammatory conditions in all of medicine due to the development and availability of highly targeted, effective topical and systemic therapies that predominantly disrupt IL-23/IL-17 cytokine signaling in affected tissues. However, future clinical studies and laboratory research are necessary to elucidate the precise cause of psoriasis as well as the underlying genetic and immune signaling pathways driving less common clinical variants and recalcitrant disease.

CT11302082_eTable.jpg

Psoriasis is a chronic inflammatory disease that affects approximately 3% of the US population.1 Plaque psoriasis comprises 80% to 90% of cases, while pustular, erythrodermic, guttate, inverse, and palmoplantar disease are less common variants (Figure 1). Psoriatic skin manifestations range from localized to widespread or generalized disease with recurrent flares. Body surface area or psoriasis area and severity index (PASI) measurements primarily focus on skin manifestations and are important for evaluating disease activity and response to treatment, but they have inherent limitations: they do not capture extracutaneous disease activity, systemic inflammation, comorbid conditions, quality of life impact, or the economic burden of psoriasis.

CT11302082_ABCD.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%20and%20B%2C%20Characteristic%20plaque%20psoriasis%20of%20the%20trunk.%20C%2C%20Inverse%20psoriasis%20involving%20the%20inframammary%20folds.%20D%2C%20Guttate%20psoriasis%20in%20an%20adult%20following%20streptococcal%20infection.%3C%2Fp%3E

A common manifestation of psoriasis is psoriatic arthritis (PsA), which can involve the nails, joints, ligaments, or tendons in 30% to 41% of affected individuals (Figure 2).2,3 A growing number of psoriasis-associated comorbidities also have been reported including metabolic syndrome4; hyperlipidemia5; cardiovascular disease6; stroke7; hypertension8; obesity9; sleep disorders10; malignancy11; infections12; inflammatory bowel disease13; and mental health disorders such as depression,14 anxiety,15 and suicidal ideation.15 Psoriatic disease also interferes with daily life activities and a patient’s overall quality of life, including interpersonal relationships, intimacy, employment, and work productivity.16 Finally, the total estimated cost of psoriasis-related health care is more than $35 billion annually,17 representing a substantial economic burden to our health care system and individual patients.

Nong_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Clinical%20manifestation%20of%20psoriatic%20arthritis%20involving%20the%20metacarpal%20joints%20of%20the%20hands.%3C%2Fp%3E

The overall burden of psoriatic disease has declined markedly in the last 2 decades due to revolutionary advances in our understanding of the immunopathogenesis of psoriasis and the subsequent development of improved therapies that predominantly interrupt IL-23/IL-17 cytokine signaling; however, critical knowledge and treatment gaps persist, underscoring the importance of ongoing clinical and research efforts in psoriatic disease. We review the working immune model of psoriasis, summarize related immune discoveries, and highlight recent therapeutic innovations that are shaping psoriatic disease management.

Current Immune Model of Psoriatic Disease

Psoriasis is an autoinflammatory T cell–mediated disease with negligible contributions from the humoral immune response. Early clinical observations reported increased inflammatory infiltrates in psoriatic skin lesions primarily consisting of both CD4+ and CD8+ T-cell populations.18,19 Additionally, patients treated with broad-acting, systemic immunosuppressive medications (eg, cyclosporine, oral corticosteroids) experienced improvement of psoriatic lesions and normalization of the immune infiltrates observed in skin biopsy specimens.20,21 These early clinical findings led to more sophisticated experimentation in xenotransplant models of psoriasis,22,23 which explored the clinical efficacy of several less immunosuppressive (eg, methotrexate, anti–tumor necrosis factor [TNF] biologics)24 or T cell–specific agents (eg, alefacept, abatacept, efalizumab).25-27 The results of these translational studies provided indisputable evidence for the role of the dysregulated immune response as the primary pathogenic process driving plaque formation; they also led to a paradigm shift in how the immunopathogenesis of psoriatic disease was viewed and paved the way for the identification and targeting of other specific proinflammatory signals produced by activated dendritic cell (DC) and T-lymphocyte populations. Among the psoriasis-associated cytokines subsequently identified and studied, elevated IL-23 and IL-17 cytokine levels in psoriatic skin were most closely associated with disease activity, and rapid normalization of IL-23/IL-17 signaling in response to effective oral or injectable antipsoriatic treatments was the hallmark of skin clearance.28 The predominant role of IL-23/IL-17 signaling in the development and maintenance of psoriatic disease is the central feature of all working immune models for this disease (Figure 3).

Nong_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Working%20immune%20model%20of%20psoriasis.%20Early%20immune%20events%20include%20activation%20of%20dendritic%20cells%20(DCs)%20and%20IL-17%E2%80%93producing%20T%20cells%20(T17)%20in%20the%20prepsoriatic%20(or%20normal-appearing)%20skin%20of%20individuals%20who%20are%20genetically%20susceptible%20and%2For%20have%20exposures%20to%20known%20psoriasis%20triggers.%20Activation%20of%20DC%20and%20T17%20populations%20in%20the%20skin%20results%20in%20increased%20production%20of%20tumor%20necrosis%20factor%20(TNF)%2C%20IL-23%2C%20and%20IL-17%20cytokines%20(namely%20IL-17A%20and%20IL-17F)%2C%20which%20work%20synergistically%20with%20other%20immune%20signals%20(IL-12%2C%20IL-22%2C%20IL-36%2C%20TNF%2C%20interferon%20%5BIFN%5D)%20to%20drive%20keratinocyte%20(KC)%20hyperproliferation.%20In%20response%20to%20upregulated%20IL-17%20signaling%2C%20substantial%20increases%20in%20keratinocyte-derived%20proteins%20(antimicrobial%20peptides%2C%20IL-19%2C%20IL-36%2C%20IL-17C)%20and%20chemotactic%20factors%20(chemokine%20%5BC-C%20motif%5D%20ligand%2020%20%5BCCL20%5D%2C%20chemokine%20%5BC-C%20motif%5D%20ligand%201%2F2%2F3%2F5%2F8%20%5BCXCL1%2F2%2F3%2F5%2F8%5D%5Bor%20IL-8%5D)%20facilitate%20further%20activation%20and%20recruitment%20of%20T17%20and%20helper%20T%20cell%20(TH1)%20lymphocytes%2C%20DCs%2C%20macrophages%2C%20and%20polymorphonuclear%20neutrophils%20(PMNs)%20into%20the%20skin.%20The%20resultant%20inflammatory%20circuit%20creates%20a%20self-amplifying%20or%20feed-forward%20immune%20response%20in%20the%20skin%20that%20leads%20to%20the%20hallmark%20clinical%20features%20of%20psoriasis%20and%20sustains%20the%20mature%20psoriatic%20plaque.%3C%2Fp%3E

Psoriasis-Associated Genetic and Environmental Risk Factors

The exact sequence of events that lead to the initiation and formation of plaque psoriasis in susceptible individuals is still poorly understood; however, several important risk factors and key immune events have been identified. First, decades of genetic research have reported more than 80 known psoriasis-associated susceptibility loci,29 which explains approximately 50% of psoriasis heritability. The major genetic determinant of psoriasis, HLA-C*06:02 (formerly HLA-Cw6), resides in the major histocompatibility complex class I region on chromosome 6p21.3 (psoriasis susceptibility gene 1, PSORS1) and is most strongly associated with psoriatic disease.30 Less common psoriasis-associated susceptibility genes also are known to directly or indirectly impact innate and adaptive immune functions that contribute to the pathogenesis of psoriasis.

Second, several nongenetic environmental risk factors for psoriasis have been reported across diverse patient populations, including skin trauma/injury, infections, alcohol/tobacco use, obesity, medication exposure (eg, lithium, antimalarials, beta-blockers), and stress.31 These genetic and/or environmental risk factors can trigger the onset of psoriatic disease at any stage of life, though most patients develop disease in early adulthood or later (age range, 50–60 years). Some patients never develop psoriasis despite exposure to environmental risk factors and/or a genetic makeup that is similar to affected first-degree relatives, which requires further study.

Prepsoriatic Skin and Initiation of Plaque Development

In response to environmental stimuli and/or other triggers of the immune system, DC and resident IL-17–producing T-cell (T17) populations become activated in predisposed individuals. Dendritic cell activation leads to the upregulation and increase of several proinflammatory cytokines, including TNF, interferon (IFN) α, IFN-γ, IL-12, and IL-23. Tumor necrosis factor and IL-23 play a vital role in psoriasis by helping to regulate the polarization and expansion of T22 and T17 cells in the skin, whereas IL-12 promotes a corresponding type 1 inflammatory response.32 Increased IL-17 and IL-22 result in alteration of the terminal differentiation and proliferative potential of epidermal keratinocytes, leading to the early clinical hallmarks of psoriatic plaques. The potential contribution of overexpressed psoriasis-related autoantigens, such as LL-37/cathelicidin, ADAMTSL5, and PLA2G4D,33 in the initiation of psoriatic plaques has been suggested but is poorly characterized.34 Whether these specific autoantigens or others presented by HLA-C variants found on antigen-presenting cells are required for the breakdown of immune tolerance and psoriatic disease initiation is highly relevant but requires further investigation and validation.

 

 

Feed-Forward Inflammation, Mature Psoriatic Plaques, and Resident Memory T Cells

In response to the upstream production of IL-23 by dermal DCs, high levels of IL-17 cytokines can be found in mature psoriatic plaques. The IL-17 family consists of 6 dimeric cytokines (IL-17A through IL-17F) that provide innate cutaneous protection against bacterial, viral, and fungal infectious agents, such as Candida albicans. Unlike other IL-17 isoforms, IL-17A and IL-17F share the same receptor complex and have the highest structural homology of any pair (approximately 50% similar).35 The relative expression of IL-17F is higher than IL-17A in psoriasis,36 though IL-17A has been considered as the predominant IL-17 cytokine found in psoriatic skin lesions due to its higher potency.

Binding of IL-17A/F with the IL-17 receptor (IL-17R) on keratinocytes contributes to the development of psoriatic plaques by inducing epidermal hyperplasia via activation of CCAAT/enhancer-binding proteins β and δ, nuclear factor κB, and signal transducer and activator of transcription 1 gene (STAT1).37,38 This also increases the expression of other keratinocyte-derived proteins (eg, human β-defensins, S-100 proteins, LL-37, other antimicrobial peptides, IL-19, IL-36, IL-17C) that act as reinforcing proinflammatory signals or chemotactic factors (eg, chemokine [C-C motif] ligand 20 [CCL20], chemokine [C-C motif] ligand 1/2/3/5 [CXCL1/2/3/5], CXCL8, IL-8) that facilitate the recruitment of additional immune cells to the skin including polymorphonuclear neutrophils (PMNs), macrophages, and DCs.39-41 Routine immunohistochemical staining for these keratinocyte-derived proteins reveals a striking epidermal gene expression gradient wherein levels of IL-17–induced proteins are most highly expressed in the uppermost layers of keratinocytes and facilitate the recruitment of immune cells into the epidermis. Activated T17 cells also stimulate the production of keratinocyte-derived chemokines (eg, CXCL9/10/11), which recruit type 1 inflammatory T-cell populations into developing psoriatic plaques.42,43 Finally, TNF, IL-36, and IL-17C cytokines act synergistically with IL-17A/F to amplify the proinflammatory effects of IL-17 signaling and further stimulate their production from T17 cell populations.40 This inflammatory circuit in the skin creates and supports a self-amplifying or positive feedback loop between the skin and immune system that commonly is referred to as feed-forward inflammation (Figure 3).34 The feed-forward inflammatory loop in psoriasis—predominantly driven by increased IL-23/IL-17 signaling—best characterizes the mature psoriatic plaque.

Several findings suggest that the influx of persistent, long-lived resident memory T cells (Trms) may contribute to the mature psoriatic plaque. It is believed that CD8+CD103+CD49a Trm cell populations may be responsible for the sharply demarcated borders of untreated psoriasis plaques or their recurrence at specific body sites such as the scalp, buttocks, extremity extensor surfaces, umbilicus, or acral skin following specific stimuli or trauma (Koebner phenomenon or isomorphic response).44,45 It is not known if repeated stimuli or trauma induce disease formation via the activation of Trm cell populations; further study in large patient cohorts is needed, but this remains an intriguing area of study for durable treatment responses and potential cures for psoriasis.

Recent Discoveries in Psoriatic Disease

Remarkable treatment outcomes for psoriasis have been achieved with multiple selective IL-17 and IL-23 inhibitors (eTable). As demonstrated in several pivotal phase 3 clinical trials for members of these classes of medications, the majority of treated psoriasis patients achieved PASI90 clearance.46 Due to their more favorable dosing schedule (ie, fewer injections) and ability to induce a durable remissionlike treatment response, IL-23 inhibitors have become the preferred treatment class for cutaneous disease, while IL-17 inhibitors may be preferred when treating patients with both plaque psoriasis and PsA.47,48 Nevertheless, the complexity of this disease is punctuated by treated patients who do not adequately respond to selective IL-23/IL-17 blockade.49 Recent and emerging treatments may shed light on these recalcitrant cases and will add to the rapidly growing arsenal of available psoriasis therapies.

The Role of IL-17F in Psoriasis and Other Inflammatory Skin Diseases

Dysregulation of IL-17A and IL-17F is associated with several chronic inflammatory conditions, such as psoriasis and PsA.35,50 Both cytokines, either as homodimers or heterodimers, can selectively bind to the heterodimeric IL-17R formed by the IL-17RA and IL-17RC subunits.35 IL-17F and IL-17C also can synergize with TNF and other cytokines to promote and support the self-sustaining inflammatory circuits in mature psoriatic plaques, though their inflammatory effects in the skin are more limited than IL-17A.51,52 Therefore, incomplete blockade of IL-17 signaling (ie, unopposed IL-17F and IL-17C) represents a potential mechanism to explain the persistence of psoriasis in patients treated with selective IL-17A inhibitors. This hypothesis is supported by reports of psoriasis patients who have inadequate clinical responses to selective IL-17A inhibition but subsequently improve with IL-17R blockade, which results in disruption of IL-17A as well as IL-17C/E/F cytokine signaling. This formed the basis for further study into the specific role of IL-17F in psoriatic disease and any potential therapeutic benefits associated with its inhibition.

Recently approved in the European Union, Canada, Australia, Japan, the United Kingdom, and the United States for moderate to severe psoriasis, bimekizumab is a novel humanized IgG antibody that selectively inhibits both IL-17A and IL-17F cytokines.53 Specifically, bimekizumab simultaneously prevents binding of IL-17A/A, IL-17A/F, and IL-17F/F dimers with the IL-17R. Compared to other IL-17 and IL-23 biologic therapies, bimekizumab (320 mg) achieved relatively higher response rates for PASI75, PASI90, and PASI100.49 Neutralization of IL-17A and IL-17F by bimekizumab also resulted in more complete suppression of cytokine responses and PMN chemotaxis than either cytokine alone in treated PsA patients,54 which is notable because of the incremental benefits of recent IL-23 and IL-17 inhibitors on inflammatory arthritis symptoms in contrast to the substantial improvements observed for cutaneous disease with those same agents.

The primary disadvantage of bimekizumab and its more complete blockade of the IL-17 signaling pathway is that treated patients have a substantially increased risk for oral candidiasis (>10%).55 However, the precise link between candidiasis and IL-17 blockade is not yet fully understood because other targeted agents that also broadly suppress IL-17 signaling (ie, IL-17R, IL-23 inhibitors) are associated with much lower rates of candidiasis.56-58 Bimekizumab also is being investigated as a novel therapy for hidradenitis suppurativa and will provide important reference information regarding the role for bispecific biologic agents in the treatment of chronic inflammatory skin diseases.59

 

 

IL-36 Signaling and Generalized Pustular Psoriasis

Recent genetic and clinical studies have expanded our understanding of the role of IL-36 signaling in the immunopathogenesis of pustular psoriasis variants. Generalized pustular psoriasis (GPP) is a rare distinct psoriasis subtype characterized by the recurrent development of widespread erythema, superficial sterile pustules, and desquamation. Systemic symptoms such as fever, malaise, itching, and skin pain accompany acute GPP flares.60 Generalized pustular psoriasis is more common in female patients (in contrast with plaque psoriasis), and acute flares may be caused by multiple stimuli including infections, hypocalcemia, initiation or discontinuation of medications (eg, oral corticosteroids), pregnancy, or stress.61,62 Flares of GPP often require emergency or in-patient care, as untreated symptoms increase the risk for severe health complications such as secondary infections, sepsis, or multisystem organ failure.63 The prevalence of GPP is estimated to be approximately 1 in 10,000 individuals in the United States,64-67 with mortality rates ranging from 0 to 3.3 deaths per 100 patient-years.67

In contrast to plaque psoriasis, aberrant IL-36 signaling is the predominant driver of GPP. IL-36 is a member of the IL-1 cytokine family that includes three IL-36 agonists (IL-36α, IL-36β, IL-36γ) and 1 endogenous antagonist (IL-36Ra, encoded by IL36RN).68 The immunopathogenesis of GPP involves dysregulation of the IL-36–chemokine–PMN axis, resulting in unopposed IL-36 signaling and the subsequent recruitment and influx of PMNs into the epidermis. IL36RN mutations are strongly associated with GPP and result in impaired function of the IL-36Ra protein, leading to unopposed IL-36 signaling.69 However, approximately two-thirds of GPP patients lack identifiable gene mutations, suggesting other immune mechanisms or triggers causing upregulated IL-36 signaling.70 In response to these triggers, increased IL-36 cytokines released by keratinocytes bind to the IL-36R, resulting in substantial keratinocyte hyperproliferation, increased IL-36 levels, and the expression of hundreds of additional inflammatory signals (eg, IL-17C, antimicrobial peptides, TNF, IL-6).71 Increased IL-36 levels also drive the production of PMN chemotactic proteins (eg, CXCL1/2/3/5/6/8 and CXCR1/2) and act synergistically with IL-17 cytokines to create an autoamplifying circuit that is analogous to the feed-forward inflammatory loop in plaque psoriasis.72 Biopsies of involved GPP skin reveal increased expression of IL-36 in the uppermost layers of the epidermis, which creates a gene expression gradient that acts as a strong attractant for PMNs and forms the basis for the hallmark pustular lesions observed in GPP patients.

Until recently, treatment strategies for GPP involved the off-label use of topical, oral, or biologic therapies approved for plaque psoriasis, which often was associated with variable or incomplete disease control. In September 2022, the US Food and Drug Administration (FDA) approved intravenous spesolimab as a first-in-class humanized monoclonal IgG1 antibody for the treatment of GPP flares in adults. Spesolimab binds to IL-36R and prevents its activation by its endogenous agonists. A phase 2, randomized, 12-week clinical trial (Effisayil-1) evaluated the efficacy and safety of a single 900-mg intravenous dose of spesolimab followed by an optional second dose 1 week later for inadequate treatment responses in 53 enrolled GPP patients (2:1 treatment to placebo randomization).73 Remarkably, more than half (19/35 [54%]) of GPP patients experienced complete resolution of pustules (GPP physician global assessment subscore of 0 [range, 0–4]) and showed sustained efficacy out to week 12 after just 1 or 2 doses of spesolimab. Overall, the safety profile of spesolimab was good; asthenia, fatigue, nausea, vomiting, headache, pruritus, infusion-related reaction and symptoms, and mild infections (eg, urinary tract infection) were the most common adverse events reported.73

Imsidolimab, a high-affinity humanized IgG4 monoclonal antibody that binds and blocks activation of IL-36R, also has completed phase 2 testing,74 with phase 3 study results expected in early 2024. The rapid onset of action and overall safety of imsidolimab was in line with and similar to spesolimab. Future approval of imsidolimab would add to the limited treatment options available for GPP and has the additional convenience of being administered to patients subcutaneously. Overall, the development of selective IL-36R inhibitors offers a much-needed therapeutic option for GPP and illustrates the importance of translational research.

Role of Tyrosine Kinase in Psoriatic Disease

The Janus kinase (JAK) enzyme family consists of 4 enzymes—tyrosine kinase 2 (TYK2), JAK1, JAK2, and JAK3—that function as intracellular transduction signals that mediate the biologic response of most extracellular cytokines and growth factors.75 Critical psoriasis-related cytokines are dependent on intact JAK-STAT signaling, including IL-23, IL-12, and type I IFNs. In 2010, a genome-wide association identified TYK2 as a psoriasis susceptibility locus,76 and loss-of-function TYK2 mutations confer a reduced risk for psoriasis.77 Unlike other JAK isoforms, TYK2 mediates biologic functions that are highly restricted to the immune responses associated with IL-23, IL-12, and type I IFN signaling.78,79 For these reasons, blockade of TYK2 signaling is an attractive therapeutic target for the potential treatment of psoriatic disease.

In September 2022, the FDA approved deucravacitinib as a first-in-class, oral, selective TYK2 inhibitor for the treatment of adult patients with moderate to severe plaque psoriasis. It was the first FDA approval of an oral small-molecule treatment for plaque psoriasis in nearly a decade. Deucravacitinib inhibits TYK2 signaling via selective binding of its unique regulatory domain, resulting in a conformational (allosteric) change that interferes with its active domain.80 This novel mechanism of action limits the unwanted blockade of other broad biologic processes mediated by JAK1/2/3. Of note, the FDA did not issue any boxed warnings for deucravacitinib as it did for other FDA-approved JAK inhibitors.

In a head-to-head, 52-week, double-blind, prospective, randomized, phase 3 study, deucravacitinib showed clear superiority over apremilast for PASI75 at week 16 (53.0% [271/511] vs 39.8% [101/254]) and week 24 (58.7% [296/504] vs 37.8% [96/254]).81 Clinical responses were sustained through week 52 and showed efficacy for difficult-to-treat areas such as the scalp, acral sites, and nails. Other advantages of deucravacitinib include once-daily dosing with no need for dose titration or adjustments for renal insufficiency as well as the absence of statistically significant differences in gastrointestinal tract symptoms compared to placebo. The most common adverse effects included nasopharyngitis, upper respiratory tract infections, headache, diarrhea, and herpes infections.81 The potential benefit of deucravacitinib for PsA and psoriasis comorbidities remains to be seen, but it is promising due to its simultaneous disruption of multiple psoriasis-related cytokine networks. Several other TYK2 inhibitors are being developed for psoriatic disease and related inflammatory conditions, underscoring the promise of targeting this intracellular pathway.

 

 

Aryl Hydrocarbon Receptor Agonism

Topical steroids are the mainstay treatment option for localized or limited plaque psoriasis due to their potent immunosuppressive effect on the skin and relatively low cost. Combined with vitamin D analogs, topical steroids result in marked improvements in disease severity and improved tolerability.82 However, chronic use of topical steroids is limited by the need for twice-daily application, resulting in poor treatment compliance; loss of efficacy over time; risk for steroid-induced skin atrophy on special body sites; and patient concerns of potential systemic effects. The discovery of novel drug targets amenable to topical inhibition is needed.

Dysregulated aryl hydrocarbon receptor (AHR) levels have been reported in atopic dermatitis and psoriasis.83 Aryl hydrocarbon receptors are ubiquitously expressed in many cell types and play an integral role in immune homeostasis within the skin, skin barrier function, protection against oxidative stressors, and regulation of proliferating melanocytes and keratinocytes.84,85 They are widely expressed in multiple immune cell types (eg, antigen-presenting cells, T lymphocytes, fibroblasts) and modulate the differentiation of T17 and T22 cells as well as their balance with regulatory T-cell populations.86 In keratinocytes, AHR helps to regulate terminal differentiation, enhance skin barrier integrity via AHR-dependent filaggrin (FLG) expression, and prevent transepidermal water loss.87,88 The mechanisms by which AHR ligands lead to the upregulation or downregulation of specific genes is intricate and highly context dependent, such as the specific ligand and cell type involved. In preclinical studies, AHR-deficient mice develop psoriasiform skin inflammation, increased IL-17 and IL-22 expression, and abnormal skin barrier function.89 Keratinocytes treated with AHR ligands in vitro modulated psoriasis-associated inflammatory cytokines, such as IL-6, IL-8, and type I and II IFNs.89,90 The use of coal tar, one of the earliest historical treatments for psoriasis, is thought to activate AHRs in the skin via organic compound mixtures containing polyaromatic hydrocarbons that help normalize the proinflammatory environment in psoriatic skin.91

In June 2022, the FDA approved tapinarof as a first-in-class, topical, nonsteroidal AHR agonist for the treatment of plaque psoriasis in adults. Although the exact mechanism of action for tapinarof has not been fully elucidated, early studies suggest that its primary function is the activation of AHR, leading to reduced T-cell expansion and T17 cell differentiation. In the imiquimod mouse model, cytokine expression of IL-17A, IL-17F, IL-19, IL-22, IL-23A, and IL-lβ in psoriasiform skin lesions were downregulated following tapinarof treatment.92 In humans, tapinarof treatment is associated with a remittive effect, in which the average time for tapinarof-treated psoriasis lesions to remain clear was approximately 4 months.93 Preliminary research investigating the mechanism by which tapinarof induces this remittive effect is ongoing and may involve the reduced activation and influx of T17 and Trm populations into the skin.94 However, these preclinical studies were performed on healthy dermatome-derived skin tissue cultured in T17-skewing conditions and needs to be replicated in larger samples sizes using human-derived psoriatic tissue. Alternatively, a strong inhibitory effect on IL-23 cytokine signaling may, in part, explain the remittive effect of tapinarof, as an analogous response is observed in patients who start and discontinue treatment with selective IL-23 antagonists. Regardless, the once-daily dosing of tapinarof and sustained treatment response is appealing to psoriasis patients. Tapinarof generally is well tolerated with mild folliculitis (>20% of patients) and contact dermatitis (5% of patients) reported as the most common skin-related adverse events.

New Roles for Phosphodiesterase 4 Inhibition

Phosphodiesterases (PDEs) are enzymes that hydrolyze cyclic nucleotides (eg, cyclic adenosine monophosphate) to regulate intracellular secondary messengers involved in the inflammatory response. One of several enzymes in the PDE family, PDE4, has been shown to have greater activity in psoriatic skin compared to healthy skin.95 Phosphodiesterase inhibitors decrease the degradation of cyclic adenosine monophosphate, which triggers protein kinase A to downregulate proinflammatory (eg, TNF-α, IL-6, IL-17, IL-12, IL-23) cytokines and increased expression of anti-inflammatory signals such as IL-10.96,97 Apremilast, the first oral PDE4 inhibitor approved by the FDA for psoriasis, offered a safe alternative to traditional oral immunosuppressive agents that had extensive risks and potential end-organ adverse effects. Unfortunately, apremilast demonstrated modest efficacy for psoriatic disease (better efficacy in the skin vs joint manifestations) and was supplanted easily by next-generation targeted biologic agents that were more efficacious and lacked the troublesome gastrointestinal tract adverse effects of PDE4 inhibition.98

Crisaborole became the first topical PDE4 inhibitor approved in the United States in December 2016 for twice-daily treatment of atopic dermatitis. Although phase 2 trial results were reported in psoriasis, this indication was never pursued, presumably due to similar improvements in primary outcome measures at week 12, compared to placebo (ClinicalTrials.gov Identifier NCT01300052).

In July 2022, the first topical PDE4 inhibitor indicated for plaque psoriasis was approved by the FDA—­roflumilast cream 0.3% for once-daily use in individuals 12 years and older. Roflumilast was found to be clinically efficacious as early as 2 weeks after its use in an early-phase clinical trial.99 In 2 phase 3 clinical trials (DERMIS-1 and DERMIS-2), roflumilast significantly increased the proportion of patients achieving PASI75 at week 8 compared to vehicle (39%–41.6% vs 5.3%–7.6%, respectively)(P<.001).100 Overall, this nonsteroidal topical therapy was found to be well tolerated, with infrequent reports of application site pain or irritation as adverse events. Similar to tapinarof, patients can apply roflumilast on all body surface areas including the face, external genitalia, and other intertriginous areas.100 Importantly, the broad immune impact of PDE4 inhibition suggests that topical roflumilast likely will be an effective treatment for several additional inflammatory conditions, including seborrheic dermatitis and atopic dermatitis, which would expand the clinical utility of this specific medication.

Conclusion

In the last 2 decades, we have witnessed a translational revolution in our understanding of the underlying genetics and immunology of psoriatic disease. Psoriasis is widely considered one of the best-managed inflammatory conditions in all of medicine due to the development and availability of highly targeted, effective topical and systemic therapies that predominantly disrupt IL-23/IL-17 cytokine signaling in affected tissues. However, future clinical studies and laboratory research are necessary to elucidate the precise cause of psoriasis as well as the underlying genetic and immune signaling pathways driving less common clinical variants and recalcitrant disease.

CT11302082_eTable.jpg

References
  1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157:940-946. doi:10.1001/jamadermatol.2021.2007
  2. Ogdie A, Langan S, Love T, et al. Prevalence and treatment patterns of psoriatic arthritis in the UK. Rheumatology (Oxford). 2013;52:568-575. doi:10.1093/rheumatology/kes324
  3. Mease PJ, Gladman DD, Papp KA, et al. Prevalence of rheumatologistdiagnosed psoriatic arthritis in patients with psoriasis in European/ North American dermatology clinics. J Am Acad Dermatol. 2013;69:729- 735. doi:10.1016/j.jaad.2013.07.023
  4. Langan SM, Seminara NM, Shin DB, et al. Prevalence of metabolic syndrome in patients with psoriasis: a population-based study in the United Kingdom. J Invest Dermatol. 2012;132(3 pt 1):556-562. doi:10.1038/jid.2011.365
  5. Wu S, Li WQ, Han J, et al. Hypercholesterolemia and risk of incident psoriasis and psoriatic arthritis in US women. Arthritis Rheumatol. 2014;66:304-310. doi:10.1002/art.38227
  6. Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and hypertension: a systematic review and meta-analysis of observational studies. J Hypertens. 2013;31:433-442; discussion 442-443. doi:10.1097/HJH.0b013e32835bcce1
  7. Gelfand JM, Dommasch ED, Shin DB, et al. The risk of stroke in patients with psoriasis. J Invest Dermatol. 2009;129:2411-2418. doi:10.1038 /jid.2009.112
  8. Armstrong AW, Lin SW, Chambers CJ, et al. Psoriasis and hypertension severity: results from a case-control study. PLoS One. 2011;6:E18227. doi:10.1371/journal.pone.0018227
  9. Naldi L, Chatenoud L, Linder D, et al. Cigarette smoking, body mass index, and stressful life events as risk factors for psoriasis: results from an Italian case-control study. J Invest Dermatol. 2005;125:61-67. doi:10.1111/j.0022-202X.2005.23681.x
  10. Yang YW, Kang JH, Lin HC. Increased risk of psoriasis following obstructive sleep apnea: a longitudinal population-based study. Sleep Med. 2012;13:285-289. doi:10.1016/j.sleep.2011.07.018
  11. Vaengebjerg S, Skov L, Egeberg A, et al. Prevalence, incidence, and risk of cancer in patients with psoriasis and psoriatic arthritis: a systematic review and meta-analysis. JAMA Dermatol. 2020;156:421-429. doi:10.1001/jamadermatol.2020.0024
  12. Loft N, Skov L, Richardson C, et al. A nationwide population-based cohort study of the incidence of severe and rare infections among adults with psoriasis in Denmark. Br J Dermatol. 2022;187:353-363. doi:10.1111/bjd.21595
  13. Fu Y, Lee CH, Chi CC. Association of psoriasis with inflammatory bowel disease: a systematic review and meta-analysis. JAMA Dermatol. 2018;154:1417-1423. doi:10.1001/jamadermatol.2018.3631
  14. Dowlatshahi EA, Wakkee M, Arends LR, et al. The prevalence and odds of depressive symptoms and clinical depression in psoriasis patients: a systematic review and meta-analysis. J Invest Dermatol. 2014;134:1542-1551. doi:10.1038/jid.2013.508
  15. Kurd SK, Troxel AB, Crits-Christoph P, et al. The risk of depression, anxiety, and suicidality in patients with psoriasis: a populationbased cohort study. Arch Dermatol. 2010;146:891-895. doi:10.1001 /archdermatol.2010.186
  16. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073-1113. doi:10.1016/j.jaad.2018.11.058
  17. Vanderpuye-Orgle J, Zhao Y, Lu J, et al. Evaluating the economic burden of psoriasis in the United States. J Am Acad Dermatol. 2015;72:961-967. doi:10.1016/j.jaad.2015.02.1099
  18. Bos JD, Hagenaars C, Das PK, et al. Predominance of “memory” T cells (CD4+, CDw29+) over “naive” T cells (CD4+, CD45R+) in both normal and diseased human skin. Arch Dermatol Res. 1989;281:24-30. doi:10.1007/BF00424268
  19. Bos JD, Hulsebosch HJ, Krieg SR, et al. Immunocompetent cells in psoriasis. in situ immunophenotyping by monoclonal antibodies. Arch Dermatol Res. 1983;275:181-189. doi:10.1007/BF00510050
  20. Griffiths CE, Powles AV, Leonard JN, et al. Clearance of psoriasis with low dose cyclosporin. Br Med J (Clin Res Ed). 1986;293:731-732. doi:10.1136/bmj.293.6549.731
  21. Ellis CN, Gorsulowsky DC, Hamilton TA, et al. Cyclosporine improves psoriasis in a double-blind study. JAMA. 1986;256:3110-3116.
  22. Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233-240. doi:10.1084/jem.20041257
  23. Ferber IA, Brocke S, Taylor-Edwards C, et al. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol. 1996;156:5-7.
  24. Bharadwaj R, Lusi CF, Mashayekh S, et al. Methotrexate suppresses psoriatic skin inflammation by inhibiting muropeptide transporter SLC46A2 activity. Immunity. 2023;56:998-1012. doi:10.1016/j. immuni.2023.04.001
  25. Jenneck C, Novak N. The safety and efficacy of alefacept in the treatment of chronic plaque psoriasis. Ther Clin Risk Manag. 2007;3:411-420.
  26. Pariser DM, Gordon KB, Papp KA, et al. Clinical efficacy of efalizumab in patients with chronic plaque psoriasis: results from three randomized placebo-controlled phase III trials: part I. J Cutan Med Surg. 2005; 9:303-312. doi:10.1007/s10227-005-0116-1
  27. Mease PJ, Gottlieb AB, van der Heijde D, et al. Efficacy and safety of abatacept, a T-cell modulator, in a randomised, double-blind, placebo-controlled, phase III study in psoriatic arthritis. Ann Rheum Dis. 2017;76:1550-1558. doi:10.1136/annrheumdis-2016-210724
  28. Hawkes JE, Yan BY, Chan TC, et al. Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis. J Immunol. 2018;201:1605-1613. doi:10.4049/jimmunol.1800013
  29. Ogawa K, Okada Y. The current landscape of psoriasis genetics in 2020. J Dermatol Sci. 2020;99:2-8. doi:10.1016/j.jdermsci.2020.05.008
  30. Arakawa A, Siewert K, Stohr J, et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med. 2015;212:2203-2212. doi:10.1084/jem.20151093
  31. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263-271. doi:10.1016/S0140-6736(07)61128-3
  32. Wang CQF, Akalu YT, Suarez-Farinas M, et al. IL-17 and TNF synergistically modulate cytokine expression while suppressing melanogenesis: potential relevance to psoriasis. J Invest Dermatol. 2013;133:2741-2752. doi:10.1038/jid.2013.237
  33. Cheung KL, Jarrett R, Subramaniam S, et la. Psoriatic T cells recognize neolipid antigens generated by mast cell phospholipase delivered by exosomes and presented by CD1a. J Exp Med. 2016;213:2399-2412. doi:10.1084/jem.20160258
  34. Hawkes JE, Gonzalez JA, Krueger JG. Autoimmunity in psoriasis: evidence for specific autoantigens. Curr Dermatol Rep. 2017;6:104-112. doi:10.1007/s13671-017-0177-6
  35. Johansen C, Usher PA, Kjellerup RB, et al. Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. Br J Dermatol. 2009;160:319-324. doi:10.1111/j.1365-2133 .2008.08902.x
  36. Kolbinger F, Loesche C, Valentin MA, et al. beta-Defensin 2 is a responsive biomarker of IL-17A-driven skin pathology in patients with psoriasis. J Allergy Clin Immunol. 2017;139:923-932. doi:10.1016/j .jaci.2016.06.038
  37. Ruddy MJ, Wong GC, Liu XK, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J Biol Chem. 2004;279:2559-2567. doi:10.1074/jbc.M308809200
  38. Shen F, Hu Z, Goswami J, et al. Identification of common transcriptional regulatory elements in interleukin-17 target genes. J Biol Chem. 2006;281:24138-24148. doi:10.1074/jbc.M604597200
  39. Harper EG, Guo C, Rizzo H, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis. J Invest Dermatol. 2009;129:2175-2183. doi:10.1038/jid.2009.65
  40. Chiricozzi A, Guttman-Yassky E, Suarez-Farinas M, et al. Integrative responses to IL-17 and TNF-alpha in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. J Invest Dermatol. 2011;131:677-687. doi:10.1038/jid.2010.340
  41. Homey B, Dieu-Nosjean MC, Wiesenborn A, et al. Up-regulation of macrophage inflammatory protein-3 alpha/CCL20 and CC chemokine receptor 6 in psoriasis. J Immunol. 2000;164:6621-6632. doi:10.4049 /jimmunol.164.12.6621
  42. Stephen-Victor E, Fickenscher H, Bayry J. IL-26: an emerging proinflammatory member of the IL-10 Cytokine family with multifaceted actions in antiviral, antimicrobial, and autoimmune responses. PLoS Pathog. 2016;12:E1005624. doi:10.1371/journal.ppat.1005624
  43. Wolk K, Witte K, Witte E, et al. IL-29 is produced by T(H)17 cells and mediates the cutaneous antiviral competence in psoriasis [published online September 25, 2013]. Sci Transl Med. doi:10.1126 /scitranslmed.3006245
  44. Kasprowicz-Furmanczyk M, Czerwinska J, Placek W, et al. Assessment of the tissue resident memory cells in lesional skin of patients with psoriasis and in healthy skin of healthy volunteers. Int J Environ Res Public Health. 2021;18:11251. doi:10.3390/ijerph182111251
  45. Cheuk S, Schlums H, Gallais Serezal I, et al. CD49a expression defines tissue-resident CD8(+) T cells poised for cytotoxic function in human skin. Immunity. 2017;46:287-300. doi:10.1016/j.immuni.2017.01.009
  46. Sawyer LM, Malottki K, Sabry-Grant C, et al. Assessing the relative efficacy of interleukin-17 and interleukin-23 targeted treatments for moderate-to-severe plaque psoriasis: a systematic review and network meta-analysis of PASI response. PLoS One. 2019;14:E0220868. doi:10.1371/journal.pone.0220868
  47. Coates LC, Kavanaugh A, Mease PJ, et al. Group for research and assessment of psoriasis and psoriatic arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68:1060-1071. doi:10.1002/art.39573
  48. Wang EA, Suzuki E, Maverakis E, et al. Targeting IL-17 in psoriatic arthritis. Eur J Rheumatol. 2017;4:272-277. doi:10.5152/eurjrheum.2017.17037
  49. Armstrong A, Fahrbach K, Leonardi C, et al. Efficacy of bimekizumab and other biologics in moderate to severe plaque psoriasis: a systematic literature review and a network meta-analysis. Dermatol Ther (Heidelb). 2022;12:1777-1792. doi:10.1007/s13555-022-00760-8
  50. van Baarsen LG, Lebre MC, van der Coelen D, et al. Heterogeneous expression pattern of interleukin 17A (IL-17A), IL-17F and their receptors in synovium of rheumatoid arthritis, psoriatic arthritis and osteoarthritis: possible explanation for nonresponse to anti-IL-17 therapy? Arthritis Res Ther. 2014;16:426. doi:10.1186/s13075-014-0426-z
  51. Hot A, Zrioual S, Toh ML, et al. IL-17A- versus IL-17F-induced intracellular signal transduction pathways and modulation by IL-17RA and IL-17RC RNA interference in rheumatoid synoviocytes. Ann Rheum Dis. 2011;70:341-348. doi:10.1136/ard.2010.132233
  52. Adams R, Maroof A, Baker T, et al. Bimekizumab, a novel humanized IgG1 antibody that neutralizes both IL-17A and IL-17F. Front Immunol. 2020;11:1894. doi:10.3389/fimmu.2020.01894
  53. Gordon KB, Foley P, Krueger JG, et al. Bimekizumab efficacy and safety in moderate to severe plaque psoriasis (BE READY): a multicentre, double-blind, placebo-controlled, randomised withdrawal phase 3 trial. Lancet. 2021;397:475-486. doi:10.1016/S0140-6736(21)00126-4
  54. Glatt S, Baeten D, Baker T, et al. Dual IL-17A and IL-17F neutralisation by bimekizumab in psoriatic arthritis: evidence from preclinical experiments and a randomised placebo-controlled clinical trial that IL-17F contributes to human chronic tissue inflammation. Ann Rheum Dis. 2018;77:523-532. doi:10.1136 /annrheumdis-2017-212127
  55. Gordon KB, Langley RG, Warren RB, et al. Bimekizumab safety in patients with moderate to severe plaque psoriasis: pooled results from phase 2 and phase 3 randomized clinical trials. JAMA Dermatol. 2022;158:735-744. doi:10.1001/jamadermatol.2022.1185
  56. Reich K, Warren RB, Lebwohl M, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med. 2021;385:142-152. doi:10.1056/NEJMoa2102383
  57. Reich K, Iversen L, Puig L, et al. Long-term efficacy and safety of brodalumab in moderate-to-severe plaque psoriasis: a post hoc pooled analysis of AMAGINE-2 and -3. J Eur Acad Dermatol Venereol. 2022;36:1275-1283. doi:10.1111/jdv.18068
  58. Papp KA, Blauvelt A, Puig L, et al. Long-term safety and efficacy of risankizumab for the treatment of moderate-to-severe plaque psoriasis: interim analysis of the LIMMitless open-label extension trial up to 5 years of follow-up. J Am Acad Dermatol. 2023;89:1149-1158. doi: 10.1016/j.jaad.2023.07.1024
  59. Glatt S, Jemec GBE, Forman S, et al. Efficacy and safety of bimekizumab in moderate to severe hidradenitis suppurativa: a phase 2, doubleblind, placebo-controlled randomized clinical trial. JAMA Dermatol. 2021;157:1279-1288. doi:10.1001/jamadermatol.2021.2905
  60. Choon SE, Lai NM, Mohammad NA, et al. Clinical profile, morbidity, and outcome of adult-onset generalized pustular psoriasis: analysis of 102 cases seen in a tertiary hospital in Johor, Malaysia. Int J Dermatol. 2014;53:676-684. doi:10.1111/ijd.12070
  61. Zheng M, Jullien D, Eyerich K. The prevalence and disease characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23 (suppl 1):5-12. doi:10.1007/s40257-021-00664-x
  62. Fujita H, Gooderham M, Romiti R. Diagnosis of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23(suppl 1):31-38. doi:10.1007/s40257-021-00652-1
  63. Choon SE, Navarini AA, Pinter A. Clinical course and characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23 (suppl 1):21-29. doi:10.1007/s40257-021-00654-z
  64. Augey F, Renaudier P, Nicolas JF. Generalized pustular psoriasis (Zumbusch): a French epidemiological survey. Eur J Dermatol. 2006;16:669-673.
  65. Ohkawara A, Yasuda H, Kobayashi H, et al. Generalized pustular psoriasis in Japan: two distinct groups formed by differences in symptoms and genetic background. Acta Derm Venereol. 1996;76:68-71. doi:10.2340/00015555766871
  66. Lee JY, Kang S, Park JS, et al. Prevalence of psoriasis in Korea: A population-based epidemiological study using the Korean National Health Insurance database. Ann Dermatol. 2017;29:761-767. doi:10.5021 /ad.2017.29.6.761
  67. Prinz JC, Choon SE, Griffiths CEM, et al. Prevalence, comorbidities and mortality of generalized pustular psoriasis: a literature review. J Eur Acad Dermatol Venereol. 2023;37:256-273. doi:10.1111/jdv.18720
  68. Johnston A, Xing X, Wolterink L, et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol. 2017;140:109-120. doi:10.1016/j.jaci.2016.08.056
  69. Rajan N, Sinclair N, Nakai H, et al. A tale of two sisters: identical IL36RN mutations and discordant phenotypes. Br J Dermatol. 2016;174:417-420. doi:10.1111/bjd.14003
  70. Ly K, Beck KM, Smith MP, et al. Diagnosis and screening of patients with generalized pustular psoriasis. Psoriasis (Auckl). 2019;9:37-42. doi:10.2147/PTT.S181808
  71. Sugiura K. Role of interleukin 36 in generalised pustular psoriasis and beyond. Dermatol Ther (Heidelb). 2022;12:315-328. doi:10.1007 /s13555-021-00677-8
  72. Akiyama M, Takeichi T, McGrath JA, et al. Autoinflammatory keratinization diseases: an emerging concept encompassing various inflammatory keratinization disorders of the skin. J Dermatol Sci. 2018;90:105-111. doi:10.1016/j.jdermsci.2018.01.012
  73. Bachelez H, Choon SE, Marrakchi S, et al. Trial of spesolimab for generalized pustular psoriasis. N Engl J Med. 2021;385:2431-2440. doi:10.1056/NEJMoa2111563
  74. Warren RB, Reich A, Kaszuba A, et al. Imsidolimab, an anti-IL-36 receptor monoclonal antibody for the treatment of generalised pustular psoriasis: results from the phase 2 GALLOP trial. Br J Dermatol. 2023;189:161-169. doi:10.1093/bjd/ljad083
  75. Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. 2017; 18:374-384. doi:10.1038/ni.3691
  76. Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case Control Consortium 2; Strange A, Capon F, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42:985-990. doi:10.1038/ng.694
  77. Enerback C, Sandin C, Lambert S, et al. The psoriasis-protective TYK2 I684S variant impairs IL-12 stimulated pSTAT4 response in skin-homing CD4+ and CD8+ memory T-cells. Sci Rep. 2018;8:7043. doi:10.1038/s41598-018-25282-2
  78. Shimoda K, Kato K, Aoki K, et al. Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function. Immunity. 2000;13:561-571. doi:10.1016/s1074-7613(00)00055-8
  79. Karaghiosoff M, Neubauer H, Lassnig C, et al. Partial impairment of cytokine responses in Tyk2-deficient mice. Immunity. 2000;13:549-560. doi:10.1016/s1074-7613(00)00054-6
  80. Burke JR, Cheng L, Gillooly KM, et al. Autoimmune pathways in mice and humans are blocked by pharmacological stabilization of the TYK2 pseudokinase domain [published online July 24, 2019]. Sci Transl Med. doi:10.1126/scitranslmed.aaw1736
  81. Strober B, Thaci D, Sofen H, et al. Deucravacitinib versus placebo and apremilast in moderate to severe plaque psoriasis: efficacy and safety results from the 52-week, randomized, double-blinded, phase 3 program for evaluation of TYK2 inhibitor psoriasis second trial. J Am Acad Dermatol. 2023;88:40-51. doi:10.1016/j.jaad.2022.08.061
  82. Stein Gold L, Lebwohl M, Menter A, et al. Aerosol foam formulation of fixed combination calcipotriene plus betamethasone dipropionate is highly efficacious in patients with psoriasis vulgaris: pooled data from three randomized controlled studies. J Drugs Dermatol. 2016;15:951-957.
  83. Beranek M, Fiala Z, Kremlacek J, et al. Serum levels of aryl hydrocarbon receptor, cytochromes p450 1a1 and 1b1 in patients with exacerbated psoriasis vulgaris. Folia Biol (Praha). 2018;64:97-102.
  84. Esser C, Rannug A. The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev. 2015;67:259- 279. doi:10.1124/pr.114.009001
  85. Furue M, Uchi H, Mitoma C, et al. Antioxidants for healthy skin: the emerging role of aryl hydrocarbon receptors and nuclear factorerythroid 2-related factor-2. Nutrients. 2017;9:223. doi:10.3390/nu9030223
  86. Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008;371:1675-1684. doi:10.1016/S0140-6736(08)60726-6
  87. Sutter CH, Olesen KM, Bhuju J, et al. AHR regulates metabolic reprogramming to promote SIRT1-dependent keratinocyte differentiation. J Invest Dermatol. 2019;139:818-826. doi:10.1016/j.jid.2018.10.019
  88. Haas K, Weighardt H, Deenen R, et al. Aryl hydrocarbon receptor in keratinocytes is essential for murine skin barrier integrity. J Invest Dermatol. 2016;136:2260-2269. doi:10.1016/j.jid.2016.06.627
  89. Di Meglio P, Duarte JH, Ahlfors H, et al. Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity. 2014;40:989-1001. doi:10.1016/j.immuni.2014.04.019
  90. Kim HO, Kim JH, Chung BY, et al. Increased expression of the aryl hydrocarbon receptor in patients with chronic inflammatory skin diseases. Exp Dermatol. 2014;23:278-281. doi:10.1111/exd.12350
  91. van den Bogaard EH, Bergboer JG, Vonk-Bergers M, et al. Coal tar induces AHR-dependent skin barrier repair in atopic dermatitis. J Clin Invest. 2013;123:917-927. doi:10.1172/JCI65642
  92. Smith SH, Jayawickreme C, Rickard DJ, et al. Tapinarof is a natural AHR agonist that resolves skin inflammation in mice and humans. J Invest Dermatol. 2017;137:2110-2119. doi:10.1016/j.jid.2017.05.004
  93. Strober B, Stein Gold L, Bissonnette R, et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J Am Acad Dermatol. 2022;87:800-806. doi:10.1016/j.jaad.2022.06.1171
  94. Mooney N, Teague JE, Gehad AE, et al. Tapinarof inhibits the formation, cytokine production, and persistence of resident memory T cells in vitro. SKIN J Cutan Med. 2023;7:S194. doi:10.25251/skin.7.supp.194
  95. Schafer PH, Truzzi F, Parton A, et al. Phosphodiesterase 4 in inflammatory diseases: effects of apremilast in psoriatic blood and in dermal myofibroblasts through the PDE4/CD271 complex. Cell Signal. 2016;28:753-763. doi:10.1016/j.cellsig.2016.01.007
  96. Li H, Zuo J, Tang W. Phosphodiesterase-4 inhibitors for the treatment of inflammatory diseases. Front Pharmacol. 2018;9:1048. doi:10.3389/ fphar.2018.01048
  97. Schafer PH, Parton A, Gandhi AK, et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br J Pharmacol. 2010;159:842-855. doi:10.1111/j.1476-5381.2009.00559.x
  98. Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol. 2015;73:37-49. doi:10.1016/j .jaad.2015.03.049
  99. Papp KA, Gooderham M, Droege M, et al. Roflumilast cream improves signs and symptoms of plaque psoriasis: results from a phase 1/2a randomized, controlled study. J Drugs Dermatol. 2020;19:734-740. doi:10.36849/JDD.2020.5370
  100. Lebwohl MG, Kircik LH, Moore AY, et al. Effect of roflumilast cream vs vehicle cream on chronic plaque psoriasis: the DERMIS-1 and DERMIS-2 randomized clinical trials. JAMA. 2022;328:1073-1084. doi:10.1001/jama.2022.15632
References
  1. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis prevalence in adults in the United States. JAMA Dermatol. 2021;157:940-946. doi:10.1001/jamadermatol.2021.2007
  2. Ogdie A, Langan S, Love T, et al. Prevalence and treatment patterns of psoriatic arthritis in the UK. Rheumatology (Oxford). 2013;52:568-575. doi:10.1093/rheumatology/kes324
  3. Mease PJ, Gladman DD, Papp KA, et al. Prevalence of rheumatologistdiagnosed psoriatic arthritis in patients with psoriasis in European/ North American dermatology clinics. J Am Acad Dermatol. 2013;69:729- 735. doi:10.1016/j.jaad.2013.07.023
  4. Langan SM, Seminara NM, Shin DB, et al. Prevalence of metabolic syndrome in patients with psoriasis: a population-based study in the United Kingdom. J Invest Dermatol. 2012;132(3 pt 1):556-562. doi:10.1038/jid.2011.365
  5. Wu S, Li WQ, Han J, et al. Hypercholesterolemia and risk of incident psoriasis and psoriatic arthritis in US women. Arthritis Rheumatol. 2014;66:304-310. doi:10.1002/art.38227
  6. Armstrong AW, Harskamp CT, Armstrong EJ. The association between psoriasis and hypertension: a systematic review and meta-analysis of observational studies. J Hypertens. 2013;31:433-442; discussion 442-443. doi:10.1097/HJH.0b013e32835bcce1
  7. Gelfand JM, Dommasch ED, Shin DB, et al. The risk of stroke in patients with psoriasis. J Invest Dermatol. 2009;129:2411-2418. doi:10.1038 /jid.2009.112
  8. Armstrong AW, Lin SW, Chambers CJ, et al. Psoriasis and hypertension severity: results from a case-control study. PLoS One. 2011;6:E18227. doi:10.1371/journal.pone.0018227
  9. Naldi L, Chatenoud L, Linder D, et al. Cigarette smoking, body mass index, and stressful life events as risk factors for psoriasis: results from an Italian case-control study. J Invest Dermatol. 2005;125:61-67. doi:10.1111/j.0022-202X.2005.23681.x
  10. Yang YW, Kang JH, Lin HC. Increased risk of psoriasis following obstructive sleep apnea: a longitudinal population-based study. Sleep Med. 2012;13:285-289. doi:10.1016/j.sleep.2011.07.018
  11. Vaengebjerg S, Skov L, Egeberg A, et al. Prevalence, incidence, and risk of cancer in patients with psoriasis and psoriatic arthritis: a systematic review and meta-analysis. JAMA Dermatol. 2020;156:421-429. doi:10.1001/jamadermatol.2020.0024
  12. Loft N, Skov L, Richardson C, et al. A nationwide population-based cohort study of the incidence of severe and rare infections among adults with psoriasis in Denmark. Br J Dermatol. 2022;187:353-363. doi:10.1111/bjd.21595
  13. Fu Y, Lee CH, Chi CC. Association of psoriasis with inflammatory bowel disease: a systematic review and meta-analysis. JAMA Dermatol. 2018;154:1417-1423. doi:10.1001/jamadermatol.2018.3631
  14. Dowlatshahi EA, Wakkee M, Arends LR, et al. The prevalence and odds of depressive symptoms and clinical depression in psoriasis patients: a systematic review and meta-analysis. J Invest Dermatol. 2014;134:1542-1551. doi:10.1038/jid.2013.508
  15. Kurd SK, Troxel AB, Crits-Christoph P, et al. The risk of depression, anxiety, and suicidality in patients with psoriasis: a populationbased cohort study. Arch Dermatol. 2010;146:891-895. doi:10.1001 /archdermatol.2010.186
  16. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80:1073-1113. doi:10.1016/j.jaad.2018.11.058
  17. Vanderpuye-Orgle J, Zhao Y, Lu J, et al. Evaluating the economic burden of psoriasis in the United States. J Am Acad Dermatol. 2015;72:961-967. doi:10.1016/j.jaad.2015.02.1099
  18. Bos JD, Hagenaars C, Das PK, et al. Predominance of “memory” T cells (CD4+, CDw29+) over “naive” T cells (CD4+, CD45R+) in both normal and diseased human skin. Arch Dermatol Res. 1989;281:24-30. doi:10.1007/BF00424268
  19. Bos JD, Hulsebosch HJ, Krieg SR, et al. Immunocompetent cells in psoriasis. in situ immunophenotyping by monoclonal antibodies. Arch Dermatol Res. 1983;275:181-189. doi:10.1007/BF00510050
  20. Griffiths CE, Powles AV, Leonard JN, et al. Clearance of psoriasis with low dose cyclosporin. Br Med J (Clin Res Ed). 1986;293:731-732. doi:10.1136/bmj.293.6549.731
  21. Ellis CN, Gorsulowsky DC, Hamilton TA, et al. Cyclosporine improves psoriasis in a double-blind study. JAMA. 1986;256:3110-3116.
  22. Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233-240. doi:10.1084/jem.20041257
  23. Ferber IA, Brocke S, Taylor-Edwards C, et al. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol. 1996;156:5-7.
  24. Bharadwaj R, Lusi CF, Mashayekh S, et al. Methotrexate suppresses psoriatic skin inflammation by inhibiting muropeptide transporter SLC46A2 activity. Immunity. 2023;56:998-1012. doi:10.1016/j. immuni.2023.04.001
  25. Jenneck C, Novak N. The safety and efficacy of alefacept in the treatment of chronic plaque psoriasis. Ther Clin Risk Manag. 2007;3:411-420.
  26. Pariser DM, Gordon KB, Papp KA, et al. Clinical efficacy of efalizumab in patients with chronic plaque psoriasis: results from three randomized placebo-controlled phase III trials: part I. J Cutan Med Surg. 2005; 9:303-312. doi:10.1007/s10227-005-0116-1
  27. Mease PJ, Gottlieb AB, van der Heijde D, et al. Efficacy and safety of abatacept, a T-cell modulator, in a randomised, double-blind, placebo-controlled, phase III study in psoriatic arthritis. Ann Rheum Dis. 2017;76:1550-1558. doi:10.1136/annrheumdis-2016-210724
  28. Hawkes JE, Yan BY, Chan TC, et al. Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis. J Immunol. 2018;201:1605-1613. doi:10.4049/jimmunol.1800013
  29. Ogawa K, Okada Y. The current landscape of psoriasis genetics in 2020. J Dermatol Sci. 2020;99:2-8. doi:10.1016/j.jdermsci.2020.05.008
  30. Arakawa A, Siewert K, Stohr J, et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med. 2015;212:2203-2212. doi:10.1084/jem.20151093
  31. Griffiths CE, Barker JN. Pathogenesis and clinical features of psoriasis. Lancet. 2007;370:263-271. doi:10.1016/S0140-6736(07)61128-3
  32. Wang CQF, Akalu YT, Suarez-Farinas M, et al. IL-17 and TNF synergistically modulate cytokine expression while suppressing melanogenesis: potential relevance to psoriasis. J Invest Dermatol. 2013;133:2741-2752. doi:10.1038/jid.2013.237
  33. Cheung KL, Jarrett R, Subramaniam S, et la. Psoriatic T cells recognize neolipid antigens generated by mast cell phospholipase delivered by exosomes and presented by CD1a. J Exp Med. 2016;213:2399-2412. doi:10.1084/jem.20160258
  34. Hawkes JE, Gonzalez JA, Krueger JG. Autoimmunity in psoriasis: evidence for specific autoantigens. Curr Dermatol Rep. 2017;6:104-112. doi:10.1007/s13671-017-0177-6
  35. Johansen C, Usher PA, Kjellerup RB, et al. Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. Br J Dermatol. 2009;160:319-324. doi:10.1111/j.1365-2133 .2008.08902.x
  36. Kolbinger F, Loesche C, Valentin MA, et al. beta-Defensin 2 is a responsive biomarker of IL-17A-driven skin pathology in patients with psoriasis. J Allergy Clin Immunol. 2017;139:923-932. doi:10.1016/j .jaci.2016.06.038
  37. Ruddy MJ, Wong GC, Liu XK, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J Biol Chem. 2004;279:2559-2567. doi:10.1074/jbc.M308809200
  38. Shen F, Hu Z, Goswami J, et al. Identification of common transcriptional regulatory elements in interleukin-17 target genes. J Biol Chem. 2006;281:24138-24148. doi:10.1074/jbc.M604597200
  39. Harper EG, Guo C, Rizzo H, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis. J Invest Dermatol. 2009;129:2175-2183. doi:10.1038/jid.2009.65
  40. Chiricozzi A, Guttman-Yassky E, Suarez-Farinas M, et al. Integrative responses to IL-17 and TNF-alpha in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. J Invest Dermatol. 2011;131:677-687. doi:10.1038/jid.2010.340
  41. Homey B, Dieu-Nosjean MC, Wiesenborn A, et al. Up-regulation of macrophage inflammatory protein-3 alpha/CCL20 and CC chemokine receptor 6 in psoriasis. J Immunol. 2000;164:6621-6632. doi:10.4049 /jimmunol.164.12.6621
  42. Stephen-Victor E, Fickenscher H, Bayry J. IL-26: an emerging proinflammatory member of the IL-10 Cytokine family with multifaceted actions in antiviral, antimicrobial, and autoimmune responses. PLoS Pathog. 2016;12:E1005624. doi:10.1371/journal.ppat.1005624
  43. Wolk K, Witte K, Witte E, et al. IL-29 is produced by T(H)17 cells and mediates the cutaneous antiviral competence in psoriasis [published online September 25, 2013]. Sci Transl Med. doi:10.1126 /scitranslmed.3006245
  44. Kasprowicz-Furmanczyk M, Czerwinska J, Placek W, et al. Assessment of the tissue resident memory cells in lesional skin of patients with psoriasis and in healthy skin of healthy volunteers. Int J Environ Res Public Health. 2021;18:11251. doi:10.3390/ijerph182111251
  45. Cheuk S, Schlums H, Gallais Serezal I, et al. CD49a expression defines tissue-resident CD8(+) T cells poised for cytotoxic function in human skin. Immunity. 2017;46:287-300. doi:10.1016/j.immuni.2017.01.009
  46. Sawyer LM, Malottki K, Sabry-Grant C, et al. Assessing the relative efficacy of interleukin-17 and interleukin-23 targeted treatments for moderate-to-severe plaque psoriasis: a systematic review and network meta-analysis of PASI response. PLoS One. 2019;14:E0220868. doi:10.1371/journal.pone.0220868
  47. Coates LC, Kavanaugh A, Mease PJ, et al. Group for research and assessment of psoriasis and psoriatic arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68:1060-1071. doi:10.1002/art.39573
  48. Wang EA, Suzuki E, Maverakis E, et al. Targeting IL-17 in psoriatic arthritis. Eur J Rheumatol. 2017;4:272-277. doi:10.5152/eurjrheum.2017.17037
  49. Armstrong A, Fahrbach K, Leonardi C, et al. Efficacy of bimekizumab and other biologics in moderate to severe plaque psoriasis: a systematic literature review and a network meta-analysis. Dermatol Ther (Heidelb). 2022;12:1777-1792. doi:10.1007/s13555-022-00760-8
  50. van Baarsen LG, Lebre MC, van der Coelen D, et al. Heterogeneous expression pattern of interleukin 17A (IL-17A), IL-17F and their receptors in synovium of rheumatoid arthritis, psoriatic arthritis and osteoarthritis: possible explanation for nonresponse to anti-IL-17 therapy? Arthritis Res Ther. 2014;16:426. doi:10.1186/s13075-014-0426-z
  51. Hot A, Zrioual S, Toh ML, et al. IL-17A- versus IL-17F-induced intracellular signal transduction pathways and modulation by IL-17RA and IL-17RC RNA interference in rheumatoid synoviocytes. Ann Rheum Dis. 2011;70:341-348. doi:10.1136/ard.2010.132233
  52. Adams R, Maroof A, Baker T, et al. Bimekizumab, a novel humanized IgG1 antibody that neutralizes both IL-17A and IL-17F. Front Immunol. 2020;11:1894. doi:10.3389/fimmu.2020.01894
  53. Gordon KB, Foley P, Krueger JG, et al. Bimekizumab efficacy and safety in moderate to severe plaque psoriasis (BE READY): a multicentre, double-blind, placebo-controlled, randomised withdrawal phase 3 trial. Lancet. 2021;397:475-486. doi:10.1016/S0140-6736(21)00126-4
  54. Glatt S, Baeten D, Baker T, et al. Dual IL-17A and IL-17F neutralisation by bimekizumab in psoriatic arthritis: evidence from preclinical experiments and a randomised placebo-controlled clinical trial that IL-17F contributes to human chronic tissue inflammation. Ann Rheum Dis. 2018;77:523-532. doi:10.1136 /annrheumdis-2017-212127
  55. Gordon KB, Langley RG, Warren RB, et al. Bimekizumab safety in patients with moderate to severe plaque psoriasis: pooled results from phase 2 and phase 3 randomized clinical trials. JAMA Dermatol. 2022;158:735-744. doi:10.1001/jamadermatol.2022.1185
  56. Reich K, Warren RB, Lebwohl M, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med. 2021;385:142-152. doi:10.1056/NEJMoa2102383
  57. Reich K, Iversen L, Puig L, et al. Long-term efficacy and safety of brodalumab in moderate-to-severe plaque psoriasis: a post hoc pooled analysis of AMAGINE-2 and -3. J Eur Acad Dermatol Venereol. 2022;36:1275-1283. doi:10.1111/jdv.18068
  58. Papp KA, Blauvelt A, Puig L, et al. Long-term safety and efficacy of risankizumab for the treatment of moderate-to-severe plaque psoriasis: interim analysis of the LIMMitless open-label extension trial up to 5 years of follow-up. J Am Acad Dermatol. 2023;89:1149-1158. doi: 10.1016/j.jaad.2023.07.1024
  59. Glatt S, Jemec GBE, Forman S, et al. Efficacy and safety of bimekizumab in moderate to severe hidradenitis suppurativa: a phase 2, doubleblind, placebo-controlled randomized clinical trial. JAMA Dermatol. 2021;157:1279-1288. doi:10.1001/jamadermatol.2021.2905
  60. Choon SE, Lai NM, Mohammad NA, et al. Clinical profile, morbidity, and outcome of adult-onset generalized pustular psoriasis: analysis of 102 cases seen in a tertiary hospital in Johor, Malaysia. Int J Dermatol. 2014;53:676-684. doi:10.1111/ijd.12070
  61. Zheng M, Jullien D, Eyerich K. The prevalence and disease characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23 (suppl 1):5-12. doi:10.1007/s40257-021-00664-x
  62. Fujita H, Gooderham M, Romiti R. Diagnosis of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23(suppl 1):31-38. doi:10.1007/s40257-021-00652-1
  63. Choon SE, Navarini AA, Pinter A. Clinical course and characteristics of generalized pustular psoriasis. Am J Clin Dermatol. 2022;23 (suppl 1):21-29. doi:10.1007/s40257-021-00654-z
  64. Augey F, Renaudier P, Nicolas JF. Generalized pustular psoriasis (Zumbusch): a French epidemiological survey. Eur J Dermatol. 2006;16:669-673.
  65. Ohkawara A, Yasuda H, Kobayashi H, et al. Generalized pustular psoriasis in Japan: two distinct groups formed by differences in symptoms and genetic background. Acta Derm Venereol. 1996;76:68-71. doi:10.2340/00015555766871
  66. Lee JY, Kang S, Park JS, et al. Prevalence of psoriasis in Korea: A population-based epidemiological study using the Korean National Health Insurance database. Ann Dermatol. 2017;29:761-767. doi:10.5021 /ad.2017.29.6.761
  67. Prinz JC, Choon SE, Griffiths CEM, et al. Prevalence, comorbidities and mortality of generalized pustular psoriasis: a literature review. J Eur Acad Dermatol Venereol. 2023;37:256-273. doi:10.1111/jdv.18720
  68. Johnston A, Xing X, Wolterink L, et al. IL-1 and IL-36 are dominant cytokines in generalized pustular psoriasis. J Allergy Clin Immunol. 2017;140:109-120. doi:10.1016/j.jaci.2016.08.056
  69. Rajan N, Sinclair N, Nakai H, et al. A tale of two sisters: identical IL36RN mutations and discordant phenotypes. Br J Dermatol. 2016;174:417-420. doi:10.1111/bjd.14003
  70. Ly K, Beck KM, Smith MP, et al. Diagnosis and screening of patients with generalized pustular psoriasis. Psoriasis (Auckl). 2019;9:37-42. doi:10.2147/PTT.S181808
  71. Sugiura K. Role of interleukin 36 in generalised pustular psoriasis and beyond. Dermatol Ther (Heidelb). 2022;12:315-328. doi:10.1007 /s13555-021-00677-8
  72. Akiyama M, Takeichi T, McGrath JA, et al. Autoinflammatory keratinization diseases: an emerging concept encompassing various inflammatory keratinization disorders of the skin. J Dermatol Sci. 2018;90:105-111. doi:10.1016/j.jdermsci.2018.01.012
  73. Bachelez H, Choon SE, Marrakchi S, et al. Trial of spesolimab for generalized pustular psoriasis. N Engl J Med. 2021;385:2431-2440. doi:10.1056/NEJMoa2111563
  74. Warren RB, Reich A, Kaszuba A, et al. Imsidolimab, an anti-IL-36 receptor monoclonal antibody for the treatment of generalised pustular psoriasis: results from the phase 2 GALLOP trial. Br J Dermatol. 2023;189:161-169. doi:10.1093/bjd/ljad083
  75. Villarino AV, Kanno Y, O’Shea JJ. Mechanisms and consequences of Jak-STAT signaling in the immune system. Nat Immunol. 2017; 18:374-384. doi:10.1038/ni.3691
  76. Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case Control Consortium 2; Strange A, Capon F, et al. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat Genet. 2010;42:985-990. doi:10.1038/ng.694
  77. Enerback C, Sandin C, Lambert S, et al. The psoriasis-protective TYK2 I684S variant impairs IL-12 stimulated pSTAT4 response in skin-homing CD4+ and CD8+ memory T-cells. Sci Rep. 2018;8:7043. doi:10.1038/s41598-018-25282-2
  78. Shimoda K, Kato K, Aoki K, et al. Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function. Immunity. 2000;13:561-571. doi:10.1016/s1074-7613(00)00055-8
  79. Karaghiosoff M, Neubauer H, Lassnig C, et al. Partial impairment of cytokine responses in Tyk2-deficient mice. Immunity. 2000;13:549-560. doi:10.1016/s1074-7613(00)00054-6
  80. Burke JR, Cheng L, Gillooly KM, et al. Autoimmune pathways in mice and humans are blocked by pharmacological stabilization of the TYK2 pseudokinase domain [published online July 24, 2019]. Sci Transl Med. doi:10.1126/scitranslmed.aaw1736
  81. Strober B, Thaci D, Sofen H, et al. Deucravacitinib versus placebo and apremilast in moderate to severe plaque psoriasis: efficacy and safety results from the 52-week, randomized, double-blinded, phase 3 program for evaluation of TYK2 inhibitor psoriasis second trial. J Am Acad Dermatol. 2023;88:40-51. doi:10.1016/j.jaad.2022.08.061
  82. Stein Gold L, Lebwohl M, Menter A, et al. Aerosol foam formulation of fixed combination calcipotriene plus betamethasone dipropionate is highly efficacious in patients with psoriasis vulgaris: pooled data from three randomized controlled studies. J Drugs Dermatol. 2016;15:951-957.
  83. Beranek M, Fiala Z, Kremlacek J, et al. Serum levels of aryl hydrocarbon receptor, cytochromes p450 1a1 and 1b1 in patients with exacerbated psoriasis vulgaris. Folia Biol (Praha). 2018;64:97-102.
  84. Esser C, Rannug A. The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacol Rev. 2015;67:259- 279. doi:10.1124/pr.114.009001
  85. Furue M, Uchi H, Mitoma C, et al. Antioxidants for healthy skin: the emerging role of aryl hydrocarbon receptors and nuclear factorerythroid 2-related factor-2. Nutrients. 2017;9:223. doi:10.3390/nu9030223
  86. Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008;371:1675-1684. doi:10.1016/S0140-6736(08)60726-6
  87. Sutter CH, Olesen KM, Bhuju J, et al. AHR regulates metabolic reprogramming to promote SIRT1-dependent keratinocyte differentiation. J Invest Dermatol. 2019;139:818-826. doi:10.1016/j.jid.2018.10.019
  88. Haas K, Weighardt H, Deenen R, et al. Aryl hydrocarbon receptor in keratinocytes is essential for murine skin barrier integrity. J Invest Dermatol. 2016;136:2260-2269. doi:10.1016/j.jid.2016.06.627
  89. Di Meglio P, Duarte JH, Ahlfors H, et al. Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditions. Immunity. 2014;40:989-1001. doi:10.1016/j.immuni.2014.04.019
  90. Kim HO, Kim JH, Chung BY, et al. Increased expression of the aryl hydrocarbon receptor in patients with chronic inflammatory skin diseases. Exp Dermatol. 2014;23:278-281. doi:10.1111/exd.12350
  91. van den Bogaard EH, Bergboer JG, Vonk-Bergers M, et al. Coal tar induces AHR-dependent skin barrier repair in atopic dermatitis. J Clin Invest. 2013;123:917-927. doi:10.1172/JCI65642
  92. Smith SH, Jayawickreme C, Rickard DJ, et al. Tapinarof is a natural AHR agonist that resolves skin inflammation in mice and humans. J Invest Dermatol. 2017;137:2110-2119. doi:10.1016/j.jid.2017.05.004
  93. Strober B, Stein Gold L, Bissonnette R, et al. One-year safety and efficacy of tapinarof cream for the treatment of plaque psoriasis: results from the PSOARING 3 trial. J Am Acad Dermatol. 2022;87:800-806. doi:10.1016/j.jaad.2022.06.1171
  94. Mooney N, Teague JE, Gehad AE, et al. Tapinarof inhibits the formation, cytokine production, and persistence of resident memory T cells in vitro. SKIN J Cutan Med. 2023;7:S194. doi:10.25251/skin.7.supp.194
  95. Schafer PH, Truzzi F, Parton A, et al. Phosphodiesterase 4 in inflammatory diseases: effects of apremilast in psoriatic blood and in dermal myofibroblasts through the PDE4/CD271 complex. Cell Signal. 2016;28:753-763. doi:10.1016/j.cellsig.2016.01.007
  96. Li H, Zuo J, Tang W. Phosphodiesterase-4 inhibitors for the treatment of inflammatory diseases. Front Pharmacol. 2018;9:1048. doi:10.3389/ fphar.2018.01048
  97. Schafer PH, Parton A, Gandhi AK, et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br J Pharmacol. 2010;159:842-855. doi:10.1111/j.1476-5381.2009.00559.x
  98. Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol. 2015;73:37-49. doi:10.1016/j .jaad.2015.03.049
  99. Papp KA, Gooderham M, Droege M, et al. Roflumilast cream improves signs and symptoms of plaque psoriasis: results from a phase 1/2a randomized, controlled study. J Drugs Dermatol. 2020;19:734-740. doi:10.36849/JDD.2020.5370
  100. Lebwohl MG, Kircik LH, Moore AY, et al. Effect of roflumilast cream vs vehicle cream on chronic plaque psoriasis: the DERMIS-1 and DERMIS-2 randomized clinical trials. JAMA. 2022;328:1073-1084. doi:10.1001/jama.2022.15632
Issue
Cutis - 113(2)
Issue
Cutis - 113(2)
Page Number
82-91,E3
Page Number
82-91,E3
Publications
Publications
Topics
Article Type
Display Headline
Expanding the Psoriasis Framework: Immunopathogenesis and Treatment Updates
Display Headline
Expanding the Psoriasis Framework: Immunopathogenesis and Treatment Updates
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Nong</fileName> <TBEID>0C02F185.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F185</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Nong</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240130T115433</firstPublished> <LastPublished>20240130T115433</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240130T115433</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Yvonne Nong, MD, MS; George Han, MD, PhD</byline> <bylineText>Yvonne Nong, MD, MS; George Han, MD, PhD; Jason E. Hawkes, MD, MS</bylineText> <bylineFull>Yvonne Nong, MD, MS; George Han, MD, PhD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>82-91,E3</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Psoriasis is a chronic inflammatory disease that affects approximately 3% of the US population.1 Plaque psoriasis comprises 80% to 90% of cases, while pustular,</metaDescription> <articlePDF>300105</articlePDF> <teaserImage/> <title>Expanding the Psoriasis Framework: Immunopathogenesis and Treatment Updates</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>February</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>2</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>psoriasis</keyword> <keyword> psoriatic arthritis</keyword> <keyword> PsA</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>February 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">281</term> <term>282</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026b7.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Expanding the Psoriasis Framework: Immunopathogenesis and Treatment Updates</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Psoriasis is a chronic heterogeneous condition with multiple available treatment options that have resulted in dramatic disease improvements for patients. IL-23/IL-17 signaling is the central immune signaling pathway driving psoriasis, though recent research has uncovered other key contributing signals such as IL-17C, IL-17F, IL-36, and tyrosine kinase 2 (TYK2). Novel therapeutic targets inhibiting these cytokines have expanded our understanding of the pathogenesis of psoriasis. IL-23/IL-17 signaling is critical for the development of epidermal hyperplasia and the mature psoriatic plaque in susceptible individuals. Increased IL-17 and IL-23 expression works synergistically with other cytokines, such as IL-12, IL-22, IL-36, tumor necrosis factor (TNF), and interferon (IFN), to help create a self-sustaining, feed-forward circuit in keratinocytes, which contributes to the chronicity of the disease. This clinical review highlights recent discoveries in the immunopathogenesis of psoriasis and summarizes new antipsoriasis therapies targeting IL-36, IL-17F, aryl hydrocarbon receptors (AHRs), phosphodiesterase 4 (PDE4), and TYK2 signaling. Despite recent success in the treatment of psoriasis, continued research is needed to further advance disease understanding and shape management strategies.</p> <p> <em><em>Cutis.</em> 2024;113:82-91, E3.</em> </p> <p>Psoriasis is a chronic inflammatory disease that affects approximately 3% of the US population.<sup>1</sup> Plaque psoriasis comprises 80% to 90% of cases, while pustular, erythrodermic, guttate, inverse, and palmoplantar disease are less common variants (Figure 1). Psoriatic skin manifestations range from localized to widespread or generalized disease with recurrent flares. Body surface area or psoriasis area and severity index (PASI) measurements primarily focus on skin manifestations and are important for evaluating disease activity and response to treatment, but they have inherent limitations: they do not capture extracutaneous disease activity, systemic inflammation, comorbid conditions, quality of life impact, or the economic burden of psoriasis.</p> <p>A common manifestation of psoriasis is psoriatic arthritis (PsA), which can involve the nails, joints, ligaments, or tendons in 30% to 41% of affected individuals (Figure 2).<sup>2,3</sup> A growing number of psoriasis-associated comorbidities also have been reported including metabolic syndrome<sup>4</sup>; hyperlipidemia<sup>5</sup>; cardiovascular disease<sup>6</sup>; stroke<sup>7</sup>; hypertension<sup>8</sup>; obesity<sup>9</sup>; sleep disorders<sup>10</sup>; malignancy<sup>11</sup>; infections<sup>12</sup>; inflammatory bowel disease<sup>13</sup>; and mental health disorders such as depression,<sup>14</sup> anxiety,<sup>15</sup> and suicidal ideation.<sup>15</sup> Psoriatic disease also interferes with daily life activities and a patient’s overall quality of life, including interpersonal relationships, intimacy, employment, and work productivity.<sup>16</sup> Finally, the total estimated cost of psoriasis-related health care is more than $35 billion annually,<sup>17</sup> representing a substantial economic burden to our health care system and individual patients. <br/><br/>The overall burden of psoriatic disease has declined markedly in the last 2 decades due to revolutionary advances in our understanding of the immunopathogenesis of psoriasis and the subsequent development of improved therapies that predominantly interrupt IL-23/IL-17 cytokine signaling; however, critical knowledge and treatment gaps persist, underscoring the importance of ongoing clinical and research efforts in psoriatic disease. We review the working immune model of psoriasis, summarize related immune discoveries, and highlight recent therapeutic innovations that are shaping psoriatic disease management. </p> <h3>Current Immune Model of Psoriatic Disease</h3> <p>Psoriasis is an autoinflammatory T cell–mediated disease with negligible contributions from the humoral immune response. Early clinical observations reported increased inflammatory infiltrates in psoriatic skin lesions primarily consisting of both CD4<span class="body"><sup>+</sup></span> and CD8<span class="body"><sup>+</sup></span> T-cell populations.<sup>18,19</sup> Additionally, patients treated with broad-acting, systemic immunosuppressive medications (eg, cyclosporine, oral corticosteroids) experienced improvement of psoriatic lesions and normalization of the immune infiltrates observed in skin biopsy specimens.<sup>20,21</sup> These early clinical findings led to more sophisticated experimentation in xenotransplant models of psoriasis,<sup>22,23</sup> which explored the clinical efficacy of several less immunosuppressive (eg, methotrexate, anti–tumor necrosis factor [TNF] biologics)<sup>24</sup> or T cell–specific agents (eg, alefacept, abatacept, efalizumab).<sup>25-27</sup> The results of these translational studies provided indisputable evidence for the role of the dysregulated immune response as the primary pathogenic process driving plaque formation; they also led to a paradigm shift in how the immunopathogenesis of psoriatic disease was viewed and paved the way for the identification and targeting of other specific proinflammatory signals produced by activated dendritic cell (DC) and T-lymphocyte populations. Among the psoriasis-associated cytokines subsequently identified and studied, elevated IL-23 and IL-17 cytokine levels in psoriatic skin were most closely associated with disease activity, and rapid normalization of IL-23/IL-17 signaling in response to effective oral or injectable antipsoriatic treatments was the hallmark of skin clearance.<sup>28</sup> The predominant role of IL-23/IL-17 signaling in the development and maintenance of psoriatic disease is the central feature of all working immune models for this disease (Figure 3).</p> <h3>Psoriasis-Associated Genetic and Environmental Risk Factors</h3> <p>The exact sequence of events that lead to the initiation and formation of plaque psoriasis in susceptible individuals is still poorly understood; however, several important risk factors and key immune events have been identified. First, decades of genetic research have reported more than 80 known psoriasis-associated susceptibility loci,<sup>29</sup> which explains approximately 50% of psoriasis heritability. The major genetic determinant of psoriasis, HLA-C*06:02 (formerly HLA-Cw6), resides in the major histocompatibility complex class I region on chromosome 6p21.3 (psoriasis susceptibility gene 1, <i>PSORS1</i>) and is most strongly associated with psoriatic disease.<sup>30</sup> Less common psoriasis-associated susceptibility genes also are known to directly or indirectly impact innate and adaptive immune functions that contribute to the pathogenesis of psoriasis. </p> <p><hl name="17870"/>Second, several nongenetic environmental risk factors for psoriasis have been reported across diverse patient populations, including skin trauma/injury, infections, alcohol/tobacco use, obesity, medication exposure (eg, lithium, antimalarials, beta-blockers), and stress.<sup>31</sup> These genetic and/or environmental risk factors can trigger the onset of psoriatic disease at any stage of life, though most patients develop disease in early adulthood or later (age range, 50–60 years). Some patients never develop psoriasis despite exposure to environmental risk factors and/or a genetic makeup that is similar to affected first-degree relatives, which requires further study.</p> <h3>Prepsoriatic Skin and Initiation of Plaque Development</h3> <p>In response to environmental stimuli and/or other triggers of the immune system, DC and resident IL-17–producing T-cell (T17) populations become activated in predisposed individuals. Dendritic cell activation leads to the upregulation and increase of several proinflammatory cytokines, including TNF, interferon (IFN) <span class="body">α,</span> IFN-<span class="body">γ</span>, IL-12, and IL-23. Tumor necrosis factor and IL-23 play a vital role in psoriasis by helping to regulate the polarization and expansion of T22 and T17 cells in the skin, whereas IL-12 promotes a corresponding type 1 inflammatory response.<sup>32</sup> Increased IL-17 and IL-22 result in alteration of the terminal differentiation and proliferative potential of epidermal keratinocytes, leading to the early clinical hallmarks of psoriatic plaques. The potential contribution of overexpressed psoriasis-related autoantigens, such as LL-37/cathelicidin, ADAMTSL5, and PLA2G4D,<sup>33</sup> in the initiation of psoriatic plaques has been suggested but is poorly characterized.<sup>34</sup> Whether these specific autoantigens or others presented by HLA-C variants found on antigen-presenting cells are required for the breakdown of immune tolerance and psoriatic disease initiation is highly relevant but requires further investigation and validation.</p> <h3>Feed-Forward Inflammation, Mature Psoriatic Plaques, and Resident Memory T Cells </h3> <p>In response to the upstream production of IL-23 by dermal DCs, high levels of IL-17 cytokines can be found in mature psoriatic plaques. The IL-17 family consists of 6 dimeric cytokines (IL-17A through IL-17F) that provide innate cutaneous protection against bacterial, viral, and fungal infectious agents, such as <i>Candida albicans. </i>Unlike other IL-17 isoforms, IL-17A and IL-17F share the same receptor complex and have the highest structural homology of any pair (approximately 50% similar).<sup>35</sup> The relative expression of IL-17F is higher than IL-17A in psoriasis,<sup>36</sup> though IL-17A has been considered as the predominant IL-17 cytokine found in psoriatic skin lesions due to its higher potency. </p> <p>Binding of IL-17A/F with the IL-17 receptor (IL-17R) on keratinocytes contributes to the development of psoriatic plaques by inducing epidermal hyperplasia via activation of CCAAT/enhancer-binding proteins <span class="body">β</span> and <span class="body">δ</span>, nuclear factor <span class="body">κ</span>B, and signal transducer and activator of transcription 1 gene (<i>STAT1</i>).<sup>37,38</sup> This also increases the expression of other keratinocyte-derived proteins (eg, human <span class="body">β</span>-defensins, S-100 proteins, LL-37, other antimicrobial peptides, IL-19, IL-36, IL-17C) that act as reinforcing proinflammatory signals or chemotactic factors (eg, chemokine [C-C motif] ligand 20 [CCL20], chemokine [C-C motif] ligand 1/2/3/5 [CXCL1/2/3/5], CXCL8, IL-8) that facilitate the recruitment of additional immune cells to the skin including polymorphonuclear neutrophils (PMNs), macrophages, and DCs.<sup>39-41</sup> Routine immunohistochemical staining for these keratinocyte-derived proteins reveals a striking epidermal gene expression gradient wherein levels of IL-17–induced proteins are most highly expressed in the uppermost layers of keratinocytes and facilitate the recruitment of immune cells into the epidermis. Activated T17 cells also stimulate the production of keratinocyte-derived chemokines (eg, CXCL9/10/11), which recruit type 1 inflammatory T-cell populations into developing psoriatic plaques.<sup>42,43</sup> Finally, TNF, IL-36, and IL-17C cytokines act synergistically with IL-17A/F to amplify the proinflammatory effects of IL-17 signaling and further stimulate their production from T17 cell populations.<sup>40</sup> This inflammatory circuit in the skin creates and supports a self-amplifying or positive feedback loop between the skin and immune system that commonly is referred to as feed-forward inflammation (Figure 3).<sup>34</sup> The feed-forward inflammatory loop in psoriasis—predominantly driven by increased IL-23/IL-17 signaling—best characterizes the mature psoriatic plaque.<br/><br/>Several findings suggest that the influx of persistent, long-lived resident memory T cells (Trms) may contribute to the mature psoriatic plaque. It is believed that CD8<span class="body"><sup>+</sup></span>CD103<span class="body"><sup>+</sup></span>CD49a<span class="body"><sup>−</sup></span> Trm cell populations may be responsible for the sharply demarcated borders of untreated psoriasis plaques or their recurrence at specific body sites such as the scalp, buttocks, extremity extensor surfaces, umbilicus, or acral skin following specific stimuli or trauma (Koebner phenomenon or isomorphic response).<sup>44,45</sup> It is not known if repeated stimuli or trauma induce disease formation via the activation of Trm cell populations; further study in large patient cohorts is needed, but this remains an intriguing area of study for durable treatment responses and potential cures for psoriasis.</p> <h3>Recent Discoveries in Psoriatic Disease</h3> <p>Remarkable treatment outcomes for psoriasis have been achieved with multiple selective IL-17 and IL-23 inhibitors (eTable). As demonstrated in several pivotal phase 3 clinical trials for members of these classes of medications, the majority of treated psoriasis patients achieved PASI90 clearance.<sup>46</sup> Due to their more favorable dosing schedule (ie, fewer injections) and ability to induce a durable remissionlike treatment response, IL-23 inhibitors have become the preferred treatment class for cutaneous disease, while IL-17 inhibitors may be preferred when treating patients with both plaque psoriasis and PsA.<sup>47,48</sup> Nevertheless, the complexity of this disease is punctuated by treated patients who do not adequately respond to selective IL-23/IL-17 blockade.<sup>49</sup> Recent and emerging treatments may shed light on these recalcitrant cases and will add to the rapidly growing arsenal of available psoriasis therapies. </p> <h3>The Role of IL-17F in Psoriasis and Other Inflammatory Skin Diseases</h3> <p>Dysregulation of IL-17A and IL-17F is associated with several chronic inflammatory conditions, such as psoriasis and PsA.<sup>35,50</sup> Both cytokines, either as homodimers or heterodimers, can selectively bind to the heterodimeric IL-17R formed by the IL-17RA and IL-17RC subunits.<sup>35</sup> IL-17F and IL-17C also can synergize with TNF and other cytokines to promote and support the self-sustaining inflammatory circuits in mature psoriatic plaques, though their inflammatory effects in the skin are more limited than IL-17A.<sup>51,52</sup> Therefore, incomplete blockade of IL-17 signaling (ie, unopposed IL-17F and IL-17C) represents a potential mechanism to explain the persistence of psoriasis in patients treated with selective IL-17A inhibitors. This hypothesis is supported by reports of psoriasis patients who have inadequate clinical responses to selective IL-17A inhibition but subsequently improve with IL-17R blockade, which results in disruption of IL-17A as well as IL-17C/E/F cytokine signaling. This formed the basis for further study into the specific role of IL-17F in psoriatic disease and any potential therapeutic benefits associated with its inhibition.</p> <p>Recently approved in the European Union, Canada, Australia, Japan, the United Kingdom, and the United States for moderate to severe psoriasis, bimekizumab is a novel humanized IgG antibody that selectively inhibits both IL-17A and IL-17F cytokines.<sup>53</sup> Specifically, bimekizumab simultaneously prevents binding of IL-17A/A, IL-17A/F, and IL-17F/F dimers with the IL-17R. Compared to other IL-17 and IL-23 biologic therapies, bimekizumab (320 mg) achieved relatively higher response rates for PASI75, PASI90, and PASI100.<sup>49</sup> Neutralization of IL-17A and IL-17F by bimekizumab also resulted in more complete suppression of cytokine responses and PMN chemotaxis than either cytokine alone in treated PsA patients,<sup>54</sup> which is notable because of the incremental benefits of recent IL-23 and IL-17 inhibitors on inflammatory arthritis symptoms in contrast to the substantial improvements observed for cutaneous disease with those same agents. <br/><br/>The primary disadvantage of bimekizumab and its more complete blockade of the IL-17 signaling pathway is that treated patients have a substantially increased risk for oral candidiasis (<span class="body">&gt;</span>10%).<sup>55</sup> However, the precise link between candidiasis and IL-17 blockade is not yet fully understood because other targeted agents that also broadly suppress IL-17 signaling (ie, IL-17R, IL-23 inhibitors) are associated with much lower rates of candidiasis.<sup>56-58</sup> Bimekizumab also is being investigated as a novel therapy for hidradenitis suppurativa and will provide important reference information regarding the role for bispecific biologic agents in the treatment of chronic inflammatory skin diseases.<sup>59</sup></p> <h3>IL-36 Signaling and Generalized Pustular Psoriasis</h3> <p>Recent genetic and clinical studies have expanded our understanding of the role of IL-36 signaling in the immunopathogenesis of pustular psoriasis variants. Generalized pustular psoriasis (GPP) is a rare distinct psoriasis subtype characterized by the recurrent development of widespread erythema, superficial sterile pustules, and desquamation. Systemic symptoms such as fever, malaise, itching, and skin pain accompany acute GPP flares.<sup>60</sup> Generalized pustular psoriasis is more common in female patients (in contrast with plaque psoriasis), and acute flares may be caused by multiple stimuli including infections, hypocalcemia, initiation or discontinuation of medications (eg, oral corticosteroids), pregnancy, or stress.<sup>61,62</sup> Flares of GPP often require emergency or in-patient care, as untreated symptoms increase the risk for severe health complications such as secondary infections, sepsis, or multisystem organ failure.<sup>63</sup> The prevalence of GPP is estimated to be approximately 1 in 10,000 individuals in the United States,<sup>64-67</sup> with mortality rates ranging from 0 to 3.3 deaths per 100 patient-years.<sup>67</sup></p> <p>In contrast to plaque psoriasis, aberrant IL-36 signaling is the predominant driver of GPP. IL-36 is a member of the IL-1 cytokine family that includes three IL-36 agonists (IL-36<span class="body">α</span>, IL-36<span class="body">β</span>, IL-36<span class="body">γ</span>) and 1 endogenous antagonist (IL-36Ra, encoded by <i>IL36RN</i>).<sup>68</sup> The immunopathogenesis of GPP involves dysregulation of the IL-36–chemokine–PMN axis, resulting in unopposed IL-36 signaling and the subsequent recruitment and influx of PMNs into the epidermis. <i>IL36RN</i> mutations are strongly associated with GPP and result in impaired function of the IL-36Ra protein, leading to unopposed IL-36 signaling.<sup>69</sup> However, approximately two-thirds of GPP patients lack identifiable gene mutations, suggesting other immune mechanisms or triggers causing upregulated IL-36 signaling.<sup>70</sup> In response to these triggers, increased IL-36 cytokines released by keratinocytes bind to the IL-36R, resulting in substantial keratinocyte hyperproliferation, increased IL-36 levels, and the expression of hundreds of additional inflammatory signals (eg, IL-17C, antimicrobial peptides, TNF, IL-6).<sup>71</sup> Increased IL-36 levels also drive the production of PMN chemotactic proteins (eg, CXCL1/2/3/5/6/8 and CXCR1/2) and act synergistically with IL-17 cytokines to create an autoamplifying circuit that is analogous to the feed-forward inflammatory loop in plaque psoriasis.<sup>72</sup> Biopsies of involved GPP skin reveal increased expression of IL-36 in the uppermost layers of the epidermis, which creates a gene expression gradient that acts as a strong attractant for PMNs and forms the basis for the hallmark pustular lesions observed in GPP patients.<br/><br/>Until recently, treatment strategies for GPP involved the off-label use of topical, oral, or biologic therapies approved for plaque psoriasis, which often was associated with variable or incomplete disease control. In September 2022, the US Food and Drug Administration (FDA) approved intravenous spesolimab as a first-in-class humanized monoclonal IgG1 antibody for the treatment of GPP flares in adults. Spesolimab binds to IL-36R and prevents its activation by its endogenous agonists. A phase 2, randomized, 12-week clinical trial (Effisayil-1) evaluated the efficacy and safety of a single 900-mg intravenous dose of spesolimab followed by an optional second dose 1 week later for inadequate treatment responses in 53 enrolled GPP patients (2:1 treatment to placebo randomization).<sup>73</sup> Remarkably, more than half (19/35 [54%]) of GPP patients experienced complete resolution of pustules (GPP physician global assessment subscore of 0 [range, 0–4]) and showed sustained efficacy out to week 12 after just 1 or 2 doses of spesolimab. Overall, the safety profile of spesolimab was good; asthenia, fatigue, nausea, vomiting, headache, pruritus, infusion-related reaction and symptoms, and mild infections (eg, urinary tract infection) were the most common adverse events reported.<sup>73</sup> <br/><br/>Imsidolimab, a high-affinity humanized IgG4 monoclonal antibody that binds and blocks activation of IL-36R, also has completed phase 2 testing,<sup>74</sup> with phase 3 study results expected in early 2024. The rapid onset of action and overall safety of imsidolimab was in line with and similar to spesolimab. Future approval of imsidolimab would add to the limited treatment options available for GPP and has the additional convenience of being administered to patients subcutaneously. Overall, the development of selective IL-36R inhibitors offers a much-needed therapeutic option for GPP and illustrates the importance of translational research.</p> <h3>Role of Tyrosine Kinase in Psoriatic Disease</h3> <p>The Janus kinase (JAK) enzyme family consists of 4 enzymes—tyrosine kinase 2 (TYK2), JAK1, JAK2, and JAK3—that function as intracellular transduction signals that mediate the biologic response of most extracellular cytokines and growth factors.<sup>75</sup> Critical psoriasis-related cytokines are dependent on intact JAK-STAT signaling, including IL-23, IL-12, and type I IFNs. In 2010, a genome-wide association identified <i>TYK2</i> as a psoriasis susceptibility locus,<sup>76</sup> and loss-of-function <i>TYK2</i> mutations confer a reduced risk for psoriasis.<sup>77</sup> Unlike other JAK isoforms, <i>TYK2</i> mediates biologic functions that are highly restricted to the immune responses associated with IL-23, IL-12, and type I IFN signaling.<sup>78,79</sup> For these reasons, blockade of <i>TYK2</i> signaling is an attractive therapeutic target for the potential treatment of psoriatic disease. </p> <p>In September 2022, the FDA approved deucravacitinib as a first-in-class, oral, selective TYK2 inhibitor for the treatment of adult patients with moderate to severe plaque psoriasis. It was the first FDA approval of an oral small-molecule treatment for plaque psoriasis in nearly a decade. Deucravacitinib inhibits TYK2 signaling via selective binding of its unique regulatory domain, resulting in a conformational (allosteric) change that interferes with its active domain.<sup>80</sup> This novel mechanism of action limits the unwanted blockade of other broad biologic processes mediated by JAK1/2/3. Of note, the FDA did not issue any boxed warnings for deucravacitinib as it did for other FDA-approved JAK inhibitors.<br/><br/>In a head-to-head, 52-week, double-blind, prospective, randomized, phase 3 study, deucravacitinib showed clear superiority over apremilast for PASI75 at week 16 (53.0% [271/511] vs 39.8% [101/254]) and week 24 (58.7% [296/504] vs 37.8% [96/254]).<sup>81</sup> Clinical responses were sustained through week 52 and showed efficacy for difficult-to-treat areas such as the scalp, acral sites, and nails. Other advantages of deucravacitinib include once-daily dosing with no need for dose titration or adjustments for renal insufficiency as well as the absence of statistically significant differences in gastrointestinal tract symptoms compared to placebo. The most common adverse effects included nasopharyngitis, upper respiratory tract infections, headache, diarrhea, and herpes infections.<sup>81</sup> The potential benefit of deucravacitinib for PsA and psoriasis comorbidities remains to be seen, but it is promising due to its simultaneous disruption of multiple psoriasis-related cytokine networks. Several other TYK2 inhibitors are being developed for psoriatic disease and related inflammatory conditions, underscoring the promise of targeting this intracellular pathway.</p> <h3>Aryl Hydrocarbon Receptor Agonism</h3> <p>Topical steroids are the mainstay treatment option for localized or limited plaque psoriasis due to their potent immunosuppressive effect on the skin and relatively low cost. Combined with vitamin D analogs, topical steroids result in marked improvements in disease severity and improved tolerability.<sup>82</sup> However, chronic use of topical steroids is limited by the need for twice-daily application, resulting in poor treatment compliance; loss of efficacy over time; risk for steroid-induced skin atrophy on special body sites; and patient concerns of potential systemic effects. The discovery of novel drug targets amenable to topical inhibition is needed.</p> <p>Dysregulated aryl hydrocarbon receptor (AHR) levels have been reported in atopic dermatitis and psoriasis.<sup>83</sup> Aryl hydrocarbon receptors are ubiquitously expressed in many cell types and play an integral role in immune homeostasis within the skin, skin barrier function, protection against oxidative stressors, and regulation of proliferating melanocytes and keratinocytes.<sup>84,85</sup> They are widely expressed in multiple immune cell types (eg, antigen-presenting cells, T lymphocytes, fibroblasts) and modulate the differentiation of T17 and T22 cells as well as their balance with regulatory T-cell populations.<sup>86</sup> In keratinocytes, AHR helps to regulate terminal differentiation, enhance skin barrier integrity via AHR-dependent filaggrin (<i>FLG</i>) expression, and prevent transepidermal water loss.<sup>87,88</sup> The mechanisms by which AHR ligands lead to the upregulation or downregulation of specific genes is intricate and highly context dependent, such as the specific ligand and cell type involved. In preclinical studies,<i> </i>AHR-deficient mice develop psoriasiform skin inflammation, increased IL-17 and IL-22 expression, and abnormal skin barrier function.<sup>89</sup><i> </i>Keratinocytes treated with AHR ligands in vitro modulated psoriasis-associated inflammatory cytokines, such as IL-6, IL-8, and type I and II IFNs.<sup>89,90</sup> The use of coal tar, one of the earliest historical treatments for psoriasis, is thought to activate AHRs in the skin via organic compound mixtures containing polyaromatic hydrocarbons that help normalize the proinflammatory environment in psoriatic skin.<sup>91</sup> <br/><br/>In June 2022, the FDA approved tapinarof as a first-in-class, topical, nonsteroidal AHR agonist for the treatment of plaque psoriasis in adults. Although the exact mechanism of action for tapinarof has not been fully elucidated, early studies suggest that its primary function is the activation of AHR, leading to reduced T-cell expansion and T17 cell differentiation.<i> </i>In the imiquimod mouse model, cytokine expression of IL-17A, IL-17F, IL-19, IL-22, IL-23A, and IL-l<span class="body">β</span> in psoriasiform skin lesions were downregulated following tapinarof treatment.<sup>92</sup> In humans, tapinarof treatment is associated with a remittive effect, in which the average time for tapinarof-treated psoriasis lesions to remain clear was approximately 4 months.<sup>93</sup> Preliminary research investigating the mechanism by which tapinarof induces this remittive effect is ongoing and may involve the reduced activation and influx of T17 and Trm populations into the skin.<sup>94</sup> However, these preclinical studies were performed on healthy dermatome-derived skin tissue cultured in T17-skewing conditions and needs to be replicated in larger samples sizes using human-derived psoriatic tissue. Alternatively, a strong inhibitory effect on IL-23 cytokine signaling may, in part, explain the remittive effect of tapinarof, as an analogous response is observed in patients who start and discontinue treatment with selective IL-23 antagonists. Regardless, the once-daily dosing of tapinarof and sustained treatment response is appealing to psoriasis patients. Tapinarof generally is well tolerated with mild folliculitis (<span class="body">&gt;</span>20% of patients) and contact dermatitis (5% of patients) reported as the most common skin-related adverse events. </p> <h3>New Roles for Phosphodiesterase 4 Inhibition</h3> <p>Phosphodiesterases (PDEs) are enzymes that hydrolyze cyclic nucleotides (eg, cyclic adenosine monophosphate) to regulate intracellular secondary messengers involved in the inflammatory response. One of several enzymes in the PDE family, PDE4, has been shown to have greater activity in psoriatic skin compared to healthy skin.<sup>95</sup> Phosphodiesterase inhibitors decrease the degradation of cyclic adenosine monophosphate, which triggers protein kinase A to downregulate proinflammatory (eg, TNF-<span class="body">α</span>, IL-6, IL-17, IL-12, IL-23) cytokines and increased expression of anti-inflammatory signals such as IL-10.<sup>96,97</sup> Apremilast, the first oral PDE4 inhibitor approved by the FDA for psoriasis, offered a safe alternative to traditional oral immunosuppressive agents that had extensive risks and potential end-organ adverse effects. Unfortunately, apremilast demonstrated modest efficacy for psoriatic disease (better efficacy in the skin vs joint manifestations) and was supplanted easily by next-generation targeted biologic agents that were more efficacious and lacked the troublesome gastrointestinal tract adverse effects of PDE4 inhibition.<sup>98</sup></p> <p>Crisaborole became the first topical PDE4 inhibitor approved in the United States in December 2016 for twice-daily treatment of atopic dermatitis. Although phase 2 trial results were reported in psoriasis, this indication was never pursued, presumably due to similar improvements in primary outcome measures at week 12, compared to placebo (ClinicalTrials.gov Identifier NCT01300052). <br/><br/>In July 2022, the first topical PDE4 inhibitor indicated for plaque psoriasis was approved by the FDA—­roflumilast cream 0.3% for once-daily use in individuals 12 years and older. Roflumilast was found to be clinically efficacious as early as 2 weeks after its use in an early-phase clinical trial.<sup>99</sup> In 2 phase 3 clinical trials (DERMIS-1 and DERMIS-2), roflumilast significantly increased the proportion of patients achieving PASI75 at week 8 compared to vehicle (39%–41.6% vs 5.3%–7.6%, respectively)(<span class="Iitalic">P</span><span class="body">&lt;</span>.001).<sup>100</sup> Overall, this nonsteroidal topical therapy was found to be well tolerated, with infrequent reports of application site pain or irritation as adverse events. Similar to tapinarof, patients can apply roflumilast on all body surface areas including the face, external genitalia, and other intertriginous areas.<sup>100</sup> Importantly, the broad immune impact of PDE4 inhibition suggests that topical roflumilast likely will be an effective treatment for several additional inflammatory conditions, including seborrheic dermatitis and atopic dermatitis, which would expand the clinical utility of this specific medication.</p> <h3>Conclusion</h3> <p>In the last 2 decades, we have witnessed a translational revolution in our understanding of the underlying genetics and immunology of psoriatic disease. Psoriasis is widely considered one of the best-managed inflammatory conditions in all of medicine due to the development and availability of highly targeted, effective topical and systemic therapies that predominantly disrupt IL-23/IL-17 cytokine signaling in affected tissues. However, future clinical studies and laboratory research are necessary to elucidate the precise cause of psoriasis as well as the underlying genetic and immune signaling pathways driving less common clinical variants and recalcitrant disease.</p> <h2>References</h2> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. Nong is from the Department of Internal Medicine, SUNY Downstate Medical Center, Brooklyn, New York. Dr. Nong also is from and Dr. Hawkes is from Integrative Skin Science and Research, Pacific Skin Institute, Sacramento, California. Dr. Han is from the Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York. </p> <p class="disclosure">Dr. Nong reports no conflict of interest. Dr. Han is or has been an investigator, consultant/advisor, or speaker for AbbVie, Amgen, Arcutis, Bausch Health, Boehringer Ingelheim, Bristol Myers Squibb, Dermavant, DermTech, Eli Lilly and Company, EPI Health, Janssen Pharmaceuticals, LEO Pharma, Novartis, Ortho Dermatologics, Pfizer Inc, Regeneron Pharmaceuticals, Sanofi Genzyme, Sun Pharmaceutical Industries Ltd, and UCB. He also has received research grants from Athenex, Bausch Health, Bond Avillion, Eli Lilly and Company, Janssen Pharmaceuticals, MC2 Therapeutics, Novartis, PellePharm, and Pfizer Inc. Dr. Hawkes is a consultant/advisor for AbbVie, Arcutis Biotherapeutics, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly and Company, Janssen Pharmaceuticals, LEO Pharma, Novartis, Pfizer, Regeneron Pharmaceuticals, Sanofi, Sun Pharmaceutical Industries Ltd, and UCB. He also is a speaker for Boehringer Ingelheim, Bristol Myers Squibb, Regeneron Pharmaceuticals, Sanofi, and UCB. <br/><br/>The eTable is in the Appendix online at www.mdedge.com/dermatology.<br/><br/>Correspondence: Jason E. Hawkes, MD, MS, Integrative Skin Science and Research, Pacific Skin Institute, 1495 River Park Dr, Sacramento, CA 95815 (hawkes3@gmail.com).doi:10.12788/cutis.0949</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Psoriasis is a chronic inflammatory condition characterized by systemic inflammation and dysregulated IL-23/IL-17 signaling.</li> <li>Modern discoveries highlight the role of additional immune signals in psoriatic disease such as IL-17C, IL-17F, IL-36, and tyrosine kinase 2, which also contribute to disease development.</li> <li>Novel systemic, oral, and topical therapies have become available and add to the rapidly growing armamentarium of safe and effective treatments for psoriatic disease.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Psoriasis is a chronic inflammatory condition characterized by systemic inflammation and dysregulated IL-23/IL-17 signaling.
  • Modern discoveries highlight the role of additional immune signals in psoriatic disease such as IL-17C, IL-17F, IL-36, and tyrosine kinase 2, which also contribute to disease development.
  • Novel systemic, oral, and topical therapies have become available and add to the rapidly growing armamentarium of safe and effective treatments for psoriatic disease.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026B7.SIG
Disable zoom
Off

Acne and Pregnancy: A Clinical Review and Practice Pearls

Article Type
Changed
Thu, 01/25/2024 - 11:53
Display Headline
Acne and Pregnancy: A Clinical Review and Practice Pearls

Acne vulgaris, or acne, is a highly common inflammatory skin disorder affecting up to 85% of the population, and it constitutes the most commonly presenting chief concern in routine dermatology practice.1 Older teenagers and young adults are most often affected by acne.2 Although acne generally is more common in males, adult-onset acne occurs more frequently in women.2,3 Black and Hispanic women are at higher risk for acne compared to those of Asian, White, or Continental Indian descent.4 As such, acne is a common concern in all women of childbearing age.

Concerns for maternal and fetal safety are important therapeutic considerations, especially because hormonal and physiologic changes in pregnancy can lead to onset of inflammatory acne lesions, particularly during the second and third trimesters.5 Female patients younger than 25 years; with a higher body mass index, prior irregular menstruation, or polycystic ovary syndrome; or those experiencing their first pregnancy are thought to be more commonly affected.5-7 In fact, acne affects up to 43% of pregnant women, and lesions typically extend beyond the face to involve the trunk.6,8-10 Importantly, one-third of women with a history of acne experience symptom relapse after disease-free periods, while two-thirds of those with ongoing disease experience symptom deterioration during pregnancy.10 Although acne is not a life-threatening condition, it has a well-documented, detrimental impact on social, emotional, and psychological well-being, namely self-perception, social interactions, quality-of-life scores, depression, and anxiety.11

Therefore, safe and effective treatment of pregnant women is of paramount importance. Because pregnant women are not included in clinical trials, there is a paucity of medication safety data, further augmented by inefficient access to available information. The US Food and Drug Administration (FDA) pregnancy safety categories were updated in 2015, letting go of the traditional A, B, C, D, and X categories.12 The Table reviews the current pregnancy classification system. In this narrative review, we summarize the most recent available data and recommendations on the safety and efficacy of acne treatment during pregnancy.

CT113001026_e_Table.jpg

Topical Treatments for Acne

Benzoyl PeroxideBenzoyl peroxide commonly is used as first-line therapy alone or in combination with other agents for the treatment of mild to moderate acne.13 It is safe for use during pregnancy.14 Although the medication is systemically absorbed, it undergoes complete metabolism to benzoic acid, a commonly used food additive.15,16 Benzoic acid has low bioavailability, as it gets rapidly metabolized by the kidneys; therefore, benzoyl peroxide is unlikely to reach clinically significant levels in the maternal circulation and consequently the fetal circulation. Additionally, it has a low risk for causing congenital malformations.17

Salicylic AcidFor mild to moderate acne, salicylic acid is a second-line agent that likely is safe for use by pregnant women at low concentrations and over limited body surface areas.14,18,19 There is minimal systemic absorption of the drug.20 Additionally, aspirin, which is broken down in the body into salicylic acid, is used in low doses for the treatment of pre-eclampsia during pregnancy.21

DapsoneThe use of dapsone gel 5% as a second-line agent has shown efficacy for mild to moderate acne.22 The oral formulation, commonly used for malaria and leprosy prophylaxis, has failed to show associated fetal toxicity or congenital anomalies.23,24 It also has been used as a first-line treatment for dermatitis herpetiformis in pregnancy.25 Although the medication likely is safe, it is better to minimize its use during the third trimester to reduce the theoretical risk for hyperbilirubinemia in the neonate.17,26-29

Azelaic AcidAzelaic acid effectively targets noninflammatory and inflammatory acne and generally is well tolerated, harboring a good safety profile.30 Topical 20% azelaic acid has localized antibacterial and comedolytic effects and is safe for use during pregnancy.31,32

 

 

Glycolic AcidLimited data exist on the safety of glycolic acid during pregnancy. In vitro studies have shown up to 27% systemic absorption depending on pH, concentration, and duration of application.33 Animal reproductive studies involving rats have shown fetal multisystem malformations and developmental abnormalities with oral administration of glycolic acid at doses far exceeding those used in humans.34 Although no human reproductive studies exist, topical glycolic acid is unlikely to reach the developing fetus in notable amounts, and the medication is likely safe for use.17,35

ClindamycinTopical clindamycin phosphate is an effective and well-tolerated agent for the treatment of mild to moderate acne.36 Its systemic absorption is minimal, and it is considered safe for use during all trimesters of pregnancy.14,17,26,27,35,37

ErythromycinTopical erythromycin is another commonly prescribed topical antibiotic used to target mild to moderate acne. However, its use recently has been associated with a decrease in efficacy secondary to the rise of antibacterial resistance in the community.38-40 Nevertheless, it remains a safe treatment for use during all trimesters of pregnancy.14,17,26,27,35,37

Topical RetinoidsVitamin A derivatives (also known as retinoids) are the mainstay for the treatment of mild to moderate acne. Limited data exist regarding pregnancy outcomes after in utero exposure.41 A rare case report suggested topical tretinoin has been associated with fetal otocerebral anomalies.42 For tazarotene, teratogenic effects were seen in animal reproductive studies at doses exceeding maximum recommended human doses.41,43 However, a large meta-analysis failed to find a clear risk for increased congenital malformations, spontaneous abortions, stillbirth, elective termination of pregnancy, low birthweight, or prematurity following first-trimester exposure to topical retinoids.44 As the level of exposure that could lead to teratogenicity in humans is unknown, avoidance of both tretinoin and tazarotene is recommended in pregnant women.41,45 Nevertheless, women inadvertently exposed should be reassured.44

Conversely, adapalene has been associated with 1 case of anophthalmia and agenesis of the optic chiasma in a fetus following exposure until 13 weeks’ gestation.46 However, a large, open-label trial prior to the patient transitioning from adapalene to over-the-counter treatment showed that the drug harbors a large and reassuring margin of safety and no risk for teratogenicity in a maximal usage trial and Pregnancy Safety Review.47 Therefore, adapalene gel 0.1% is a safe and effective medication for the treatment of acne in a nonprescription environment and does not pose harm to the fetus.

ClascoteroneClascoterone is a novel topical antiandrogenic drug approved for the treatment of hormonal and inflammatory moderate to severe acne.48-51 Human reproductive data are limited to 1 case of pregnancy that occurred during phase 3 trial investigations, and no adverse outcomes were reported.51 Minimal systemic absorption follows topical use.52 Nonetheless, dose-independent malformations were reported in animal reproductive studies.53 As such, it remains better to avoid the use of clascoterone during pregnancy pending further safety data.

Minocycline FoamMinocycline foam 4% is approved to treat inflammatory lesions of nonnodular moderate to severe acne in patients 9 years and older.54 Systemic absorption is minimal, and the drug has limited bioavailability with minimal systemic accumulation in the patient’s serum.55 Given this information, it is unlikely that topical minocycline will reach notable levels in the fetal serum or harbor teratogenic effects, as seen with the oral formulation.56 However, it may be best to avoid its use during the second and third trimesters given the potential risk for tooth discoloration in the fetus.57,58

 

 

Systemic Treatments for Acne

IsotretinoinIsotretinoin is the most effective treatment for moderate to severe acne with a well-documented potential for long-term clearance.59 Its use during pregnancy is absolutely contraindicated, as the medication is a well-known teratogen. Associated congenital malformations include numerous craniofacial defects, cardiovascular and neurologic malformations, or thymic disorders that are estimated to affect 20% to 35% of infants exposed in utero.60 Furthermore, strict contraception use during treatment is mandated for patients who can become pregnant. It is recommended to wait at least 1 month and 1 menstrual cycle after medication discontinuation before attempting to conceive.17 Pregnancy termination is recommended if conception occurs during treatment with isotretinoin.

SpironolactoneSpironolactone is an androgen-receptor antagonist commonly prescribed off label for mild to severe acne in females.61,62 Spironolactone promotes the feminization of male fetuses and should be avoided in pregnancy.63

Doxycycline/MinocyclineTetracyclines are the most commonly prescribed oral antibiotics for moderate to severe acne.64 Although highly effective at treating acne, tetracyclines generally should be avoided in pregnancy. First-trimester use of doxycycline is not absolutely contraindicated but should be reserved for severe illness and not employed for the treatment of acne. However, accidental exposure to doxycycline has not been associated with congenital malformations.65 Nevertheless, after the 15th week of gestation, permanent tooth discoloration and bone growth inhibition in the fetus are serious and well-documented risks.14,17 Additional adverse events following in utero exposure include infantile inguinal hernia, hypospadias, and limb hypoplasia.63

SarecyclineSarecycline is a novel tetracycline-class antibiotic for the treatment of moderate to severe inflammatory acne. It has a narrower spectrum of activity compared to its counterparts within its class, which translates to an improved safety profile, namely when it comes to gastrointestinal tract microbiome disruption and potentially decreased likelihood of developing bacterial resistance.66 Data on human reproductive studies are limited, but it is advisable to avoid sarecycline in pregnancy, as it may cause adverse developmental effects in the fetus, such as reduced bone growth, in addition to the well-known tetracycline-associated risk for permanent discoloration of the teeth if used during the second and third trimesters.67,68

ErythromycinOral erythromycin targets moderate to severe inflammatory acne and is considered safe for use during pregnancy.69,70 There has been 1 study reporting an increased risk for atrial and ventricular septal defects (1.8%) and pyloric stenosis (0.2%), but these risks are still uncertain, and erythromycin is considered compatible with pregnancy.71 However, erythromycin estolate formulations should be avoided given the associated 10% to 15% risk for reversible cholestatic liver injury.72 Erythromycin base or erythromycin ethylsuccinate formulations should be favored.

Systemic SteroidsPrednisone is indicated for severe acne with scarring and should only be used during pregnancy after clearance from the patient’s obstetrician. Doses of 0.5 mg/kg or less should be prescribed in combination with systemic antibiotics as well as agents for bone and gastrointestinal tract prophylaxis.29

ZincThe exact mechanism by which zinc exerts its effects to improve acne remains largely obscure. It has been found effective against inflammatory lesions of mild to moderate acne.73 Generally recommended dosages range from 30 to 200 mg/d but may be associated with gastrointestinal tract disturbances. Dosages of 75 mg/d have shown no harm to the fetus.74 When taking this supplement, patients should not exceed the recommended doses given the risk for hypocupremia associated with high-dose zinc supplementation.

 

 

Light-Based Therapies

PhototherapyNarrowband UVB phototherapy is effective for the treatment of mild to moderate acne.75 It has been proven to be a safe treatment option during pregnancy, but its use has been associated with decreased folic acid levels.76-79 Therefore, in addition to attaining baseline folic acid serum levels, supplementation with folic acid prior to treatment, as per routine prenatal guidelines, should be sought.80

AviClearThe AviClear (Cutera) laser is the first device cleared by the FDA for mild to severe acne in March 2022.81 The FDA clearance for the Accure (Accure Acne Inc) laser, also targeting mild to severe acne, followed soon after (November 2022). Both lasers harbor a wavelength of 1726 nm and target sebaceous glands with electrothermolysis.82,83 Further research and long-term safety data are required before using them in pregnancy.

Other Therapies

Cosmetic PeelsGlycolic acid peels induce epidermolysis and desquamation.84 Although data on use during pregnancy are limited, these peels have limited dermal penetration and are considered safe for use in pregnancy.33,85,86 Similarly, keratolytic lactic acid peels harbor limited dermal penetration and can be safely used in pregnant women.87-89 Salicylic acid peels also work through epidermolysis and desquamation84; however, they tend to penetrate deeper into the skin, reaching down to the basal layer, if large areas are treated or when applied under occlusion.86,90 Although their use is not contraindicated in pregnancy, they should be limited to small areas of coverage.91

Intralesional TriamcinoloneAcne cysts and inflammatory papules can be treated with intralesional triamcinolone injections to relieve acute symptoms such as pain.92 Low doses at concentrations of 2.5 mg/mL are considered compatible with pregnancy when indicated.29

Approaching the Patient Clinical Encounter

In patients seeking treatment prior to conception, a few recommendations can be made to minimize the risk for acne recurrence or flares during pregnancy. For instance, because data show an association between increased acne severity in those with a higher body mass index and in pregnancy, weight loss may be recommended prior to pregnancy to help mitigate symptoms after conception.7 The Figure summarizes our recommendations for approaching and treating acne in pregnancy.

ret_Yaghi_figure.jpg
%3Cp%3EAn%20algorithm-based%20approach%20for%20the%20management%20of%20acne%20during%20pregnancy.%3C%2Fp%3E

In all patients, grading the severity of the patient’s acne as mild, moderate, or severe is the first step. The presence of scarring is an additional consideration during the physical examination and should be documented. A careful discussion of treatment expectations and prognosis should be the focus before treatment initiation. Meticulous documentation of the physical examination and discussion with the patient should be prioritized.

To minimize toxicity and risks to the developing fetus, monotherapy is favored. Topical therapy should be considered first line. Safe regimens include mild nonabrasive washes, such as those containing benzoyl peroxide or glycolic acid, or topical azelaic acid or clindamycin phosphate for mild to moderate acne. More severe cases warrant the consideration of systemic medications as second line, as more severe acne is better treated with oral antibiotics such as the macrolides erythromycin or clindamycin or systemic corticosteroids when concern exists for severe scarring. The additional use of physical sunscreen also is recommended.

An important topic to address during the clinical encounter is cautious intake of oral supplements for acne during pregnancy, as they may contain harmful and teratogenic ingredients. A recent search focusing on acne supplements available online between March and May 2020 uncovered 49 different supplements, 26 (53%) of which contained vitamin A.93 Importantly, 3 (6%) of these 49 supplements were likely teratogenic, 4 (8%) contained vitamin A doses exceeding the recommended daily nutritional intake level, and 15 (31%) harbored an unknown teratogenic risk. Furthermore, among the 6 (12%) supplements with vitamin A levels exceeding 10,000 IU, 2 lacked any mention of pregnancy warning, including the supplement with the highest vitamin A dose found in this study.93 Because dietary supplements are not subject to the same stringent regulations by the FDA as drugs, inadvertent use by unaware patients ought to be prevented by careful counseling and education.

Finally, patients should be counseled to seek care following delivery for potentially updated medication management of acne, especially if they are breastfeeding. Co-management with a pediatrician may be indicated during lactation, particularly when newborns are born preterm or with other health conditions that may warrant additional caution with the use of certain agents.

References
  1. Bhate K, Williams H. Epidemiology of acne vulgaris. Br J Dermatol. 2013;168:474-485.
  2. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020;10:5754.
  3. Fisk WA, Lev-Tov HA, Sivamani RK. Epidemiology and management of acne in adult women. Curr Dermatol Rep. 2014;3:29-39.
  4. Perkins A, Cheng C, Hillebrand G, et al. Comparison of the epidemiology of acne vulgaris among Caucasian, Asian, Continental Indian and African American women. J Eur Acad Dermatol Venereol. 2011;25:1054-1060.
  5. Yang CC, Huang YT, Yu CH, et al. Inflammatory facial acne during uncomplicated pregnancy and post‐partum in adult women: a preliminary hospital‐based prospective observational study of 35 cases from Taiwan. J Eur Acad Dermatol Venereol. 2016;30:1787-1789.
  6. Dréno B, Blouin E, Moyse D, et al. Acne in pregnant women: a French survey. Acta Derm Venereol. 2014;94:82-83.
  7. Kutlu Ö, Karadag˘ AS, Ünal E, et al. Acne in pregnancy: a prospective multicenter, cross‐sectional study of 295 patients in Turkey. Int J Dermatol. 2020;59:1098-1105.
  8. Hoefel IDR, Weber MB, Manzoni APD, et al. Striae gravidarum, acne, facial spots, and hair disorders: risk factors in a study with 1284 puerperal patients. J Pregnancy. 2020;2020:8036109.
  9. Ayanlowo OO, Otrofanowei E, Shorunmu TO, et al. Pregnancy dermatoses: a study of patients attending the antenatal clinic at two tertiary care centers in south west Nigeria. PAMJ Clin Med. 2020;3.
  10. Bechstein S, Ochsendorf F. Acne and rosacea in pregnancy. Hautarzt. 2017;68:111-119.
  11. Habeshian KA, Cohen BA. Current issues in the treatment of acne vulgaris. Pediatrics. 2020;145(suppl 2):S225-S230.
  12. Content and format of labeling for human prescription drug and biological products; requirements for pregnancy and lactation labeling (21 CFR 201). Fed Regist. 2014;79:72064-72103.
  13. Sagransky M, Yentzer BA, Feldman SR. Benzoyl peroxide: a review of its current use in the treatment of acne vulgaris. Expert Opin Pharmacother. 2009;10:2555-2562.
  14. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation: part I. Pregnancy. J Am Acad Dermatol. 2014;70:401.e1-401.e14; quiz 415.
  15. Wolverton SE. Systemic corticosteroids. Comprehensive Dermatol Drug Ther. 2012;3:143-168.
  16. Kirtschig G, Schaefer C. Dermatological medications and local therapeutics. In: Schaefer C, Peters P, Miller RK, eds. Drugs During Pregnancy and Lactation. 3rd edition. Elsevier; 2015:467-492.
  17. Pugashetti R, Shinkai K. Treatment of acne vulgaris in pregnant patients. Dermatol Ther. 2013;26:302-311.
  18. Touitou E, Godin B, Shumilov M, et al. Efficacy and tolerability of clindamycin phosphate and salicylic acid gel in the treatment of mild to moderate acne vulgaris. J Eur Acad Dermatol Venereol. 2008;22:629-631.
  19. Schaefer C, Peters PW, Miller RK, eds. Drugs During Pregnancy and Lactation: Treatment Options and Risk Assessment. 2nd ed. Academic Press; 2014.
  20. Birmingham B, Greene D, Rhodes C. Systemic absorption of topical salicylic acid. Int J Dermatol. 1979;18:228-231.
  21. Trivedi NA. A meta-analysis of low-dose aspirin for prevention of preeclampsia. J Postgrad Med. 2011;57:91-95.
  22. Lucky AW, Maloney JM, Roberts J, et al. Dapsone gel 5% for the treatment of acne vulgaris: safety and efficacy of long-term (1 year) treatment. J Drugs Dermatol. 2007;6:981-987.
  23. Nosten F, McGready R, d’Alessandro U, et al. Antimalarial drugs in pregnancy: a review. Curr Drug Saf. 2006;1:1-15.
  24. Brabin BJ, Eggelte TA, Parise M, et al. Dapsone therapy for malaria during pregnancy: maternal and fetal outcomes. Drug Saf. 2004;27:633-648.
  25. Tuffanelli DL. Successful pregnancy in a patient with dermatitis herpetiformis treated with low-dose dapsone. Arch Dermatol. 1982;118:876.
  26. Meredith FM, Ormerod AD. The management of acne vulgaris in pregnancy. Am J Clin Dermatol. 2013;14:351-358.
  27. Kong Y, Tey H. Treatment of acne vulgaris during pregnancy and lactation. Drugs. 2013;73:779-787.
  28. Leachman SA, Reed BR. The use of dermatologic drugs in pregnancy and lactation. Dermatol Clin. 2006;24:167-197.
  29. Ly S, Kamal K, Manjaly P, et al. Treatment of acne vulgaris during pregnancy and lactation: a narrative review. Dermatol Ther. 2023;13:115-130.
  30. Webster G. Combination azelaic acid therapy for acne vulgaris. J Am Acad Dermatol. 2000;43:S47-S50.
  31. Archer CB, Cohen SN, Baron SE. Guidance on the diagnosis and clinical management of acne. Clin Exp Dermatol. 2012;37(suppl 1):1-6.
  32. Graupe K, Cunliffe W, Gollnick H, et al. Efficacy and safety of topical azelaic acid (20 percent cream): an overview of results from European clinical trials and experimental reports. Cutis. 1996;57(1 suppl):20-35.
  33. Bozzo P, Chua-Gocheco A, Einarson A. Safety of skin care products during pregnancy. Can Fam Physician. 2011;57:665-667.
  34. Munley SM, Kennedy GL, Hurtt ME. Developmental toxicity study of glycolic acid in rats. Drug Chem Toxicol. 1999;22:569-582.
  35. Chien AL, Qi J, Rainer B, et al. Treatment of acne in pregnancy. J Am Board Fam Med. 2016;29:254-262.
  36. Stuart B, Maund E, Wilcox C, et al. Topical preparations for the treatment of mild‐to‐moderate acne vulgaris: systematic review and network meta‐analysis. Br J Dermatol. 2021;185:512-525.
  37. van Hoogdalem EJ, Baven TL, Spiegel‐Melsen I, et al. Transdermal absorption of clindamycin and tretinoin from topically applied anti‐acne formulations in man. Biopharm Drug Dispos. 1998;19:563-569.
  38. Austin BA, Fleischer AB Jr. The extinction of topical erythromycin therapy for acne vulgaris and concern for the future of topical clindamycin. J Dermatolog Treat. 2017;28:145-148.
  39. Eady EA, Cove J, Holland K, et al. Erythromycin resistant propionibacteria in antibiotic treated acne patients: association with therapeutic failure. Br J. Dermatol. 1989;121:51-57.
  40. Alkhawaja E, Hammadi S, Abdelmalek M, et al. Antibiotic resistant Cutibacterium acnes among acne patients in Jordan: a cross sectional study. BMC Dermatol. 2020;20:1-9.
  41. Han G, Wu JJ, Del Rosso JQ. Use of topical tazarotene for the treatment of acne vulgaris in pregnancy: a literature review. J Clin Aesthet Dermatol. 2020;13:E59-E65.
  42. Selcen D, Seidman S, Nigro MA. Otocerebral anomalies associated with topical tretinoin use. Brain Dev. 2000;22:218-220.
  43. Moretz D. Drug Class Update with New Drug Evaluations: Topical Products for Inflammatory Skin Conditions. Oregon State University Drug Use & Research Management Program; December 2022. Accessed January 8, 2024. https://www.orpdl.org/durm/meetings/meetingdocs/2022_12_01/archives/2022_12_01_Inflammatory_Skin_Dz_ClassUpdate.pdf
  44. Kaplan YC, Ozsarfati J, Etwel F, et al. Pregnancy outcomes following first‐trimester exposure to topical retinoids: a systematic review and meta‐analysis. Br J Dermatol. 2015;173:1132-1141.
  45. Menter A. Pharmacokinetics and safety of tazarotene. J Am Acad Dermatol. 2000;43(2, pt 3):S31-S35.
  46. Autret E, Berjot M, Jonville-Béra A-P, et al. Anophthalmia and agenesis of optic chiasma associated with adapalene gel in early pregnancy. Lancet. 1997;350:339.
  47. Weiss J, Mallavalli S, Meckfessel M, et al. Safe use of adapalene 0.1% gel in a non-prescription environment. J Drugs Dermatol. 2021;20:1330-1335.
  48. Alessandro Mazzetti M. A phase 2b, randomized, double-blind vehicle controlled, dose escalation study evaluating clascoterone 0.1%, 0.5%, and 1% topical cream in subjects with facial acne. J Drugs Dermatol. 2019;18:570-575.
  49. Eichenfield L, Hebert A, Gold LS, et al. Open-label, long-term extension study to evaluate the safety of clascoterone (CB-03-01) cream, 1% twice daily, in patients with acne vulgaris. J Am Acad Dermatol. 2020;83:477-485.
  50. Trifu V, Tiplica GS, Naumescu E, et al. Cortexolone 17α‐propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. a pilot randomized, double‐blind comparative study vs. placebo and tretinoin 0.05% cream. Br J Dermatol. 2011;165:177-183.
  51. Hebert A, Thiboutot D, Gold LS, et al. Efficacy and safety of topical clascoterone cream, 1%, for treatment in patients with facial acne: two phase 3 randomized clinical trials. JAMA Dermatol. 2020;156:621-630.
  52. Alkhodaidi ST, Al Hawsawi KA, Alkhudaidi IT, et al. Efficacy and safety of topical clascoterone cream for treatment of acne vulgaris: a systematic review and meta‐analysis of randomized placebo‐controlled trials. Dermatol Ther. 2021;34:e14609.
  53. Clasoterone. Package insert. Cassiopea Inc; 2020.
  54. Paik J. Topical minocycline foam 4%: a review in acne vulgaris. Am J Clin Dermatol. 2020;21:449-456.
  55. Jones TM, Ellman H. Pharmacokinetic comparison of once-daily topical minocycline foam 4% vs oral minocycline for moderate-to-severe acne. J Drugs Dermatol. 2017;16:1022-1028.
  56. Minocycline hydrochloride extended-release tablets. Package insert. JG Pharma; July 2020. Accessed January 8, 2024. https://www.jgpharmainc.com/assets/pdf/minocycline-hydrochloride.pdf
  57. Dinnendahl V, Fricke U (eds). Arzneistoff-Profile: Basisinformation über arzneiliche Wirkstoffe. Govi Pharmazeutischer Verlag; 2010.
  58. Martins AM, Marto JM, Johnson JL, et al. A review of systemic minocycline side effects and topical minocycline as a safer alternative for treating acne and rosacea. Antibiotics. 2021;10:757.
  59. Landis MN. Optimizing isotretinoin treatment of acne: update on current recommendations for monitoring, dosing, safety, adverse effects, compliance, and outcomes. Am J Clin Dermatol. 2020;21:411-419.
  60. Draghici C-C, Miulescu R-G, Petca R-C, et al. Teratogenic effect of isotretinoin in both fertile females and males. Exp Ther Med. 2021;21:1-5.
  61. Barker RA, Wilcox C, Layton AM. Oral spironolactone for acne vulgaris in adult females: an update of the literature. Am J Clin Dermatol. 2020;21:303-305.
  62. Han JJ, Faletsky A, Barbieri JS, et al. New acne therapies and updates on use of spironolactone and isotretinoin: a narrative review. Dermatol Ther (Heidelb). 2021;11:79-91.
  63. Briggs GG, Freeman RK, Yaffe SJ. Drugs in Pregnancy and Lactation: A Reference Guide to Fetal and Neonatal Risk. Lippincott Williams & Wilkins; 2012.
  64. Patel DJ, Bhatia N. Oral antibiotics for acne. Am J Clin Dermatol. 2021;22:193-204.
  65. Jick H, Holmes LB, Hunter JR, et al. First-trimester drug use and congenital disorders. JAMA. 1981;246:343-346.
  66. Valente Duarte de Sousa IC. An overview of sarecycline for the treatment of moderate-to-severe acne vulgaris. Exp Opin Pharmacother. 2021;22:145-154.
  67. Hussar DA, Chahine EB. Omadacycline tosylate, sarecycline hydrochloride, rifamycin sodium, and moxidectin. J Am Pharm Assoc. 2019;59:756-760.
  68. Haidari W, Bruinsma R, Cardenas-de la Garza JA, et al. Sarecycline review. Ann Pharmacother. 2020;54:164-170.
  69. Feldman S, Careccia RE, Barham KL, et al. Diagnosis and treatment of acne. Am Fam Physician. 2004;69:2123-2130.
  70. Gammon WR, Meyer C, Lantis S, et al. Comparative efficacy of oral erythromycin versus oral tetracycline in the treatment of acne vulgaris: a double-blind study. J Am Acad Dermatol. 1986;14:183-186.
  71. Källén BA, Olausson PO, Danielsson BR. Is erythromycin therapy teratogenic in humans? Reprod Toxicol. 2005;20:209-214.
  72. McCormack WM, George H, Donner A, et al. Hepatotoxicity of erythromycin estolate during pregnancy. Antimicrob Agents Chemother. 1977;12:630-635.
  73. Cervantes J, Eber AE, Perper M, et al. The role of zinc in the treatment of acne: a review of the literature. Dermatolog Ther. 2018;31:e12576.
  74. Dréno B, Blouin E. Acne, pregnant women and zinc salts: a literature review [in French]. Ann Dermatol Venereol. 2008;135:27-33.
  75. Eid MM, Saleh MS, Allam NM, et al. Narrow band ultraviolet B versus red light-emitting diodes in the treatment of facial acne vulgaris: a randomized controlled trial. Photobiomodul Photomed Laser Surg. 2021;39:418-424.
  76. Zeichner JA. Narrowband UV-B phototherapy for the treatment of acne vulgaris during pregnancy. Arch Dermatol. 2011;147:537-539.
  77. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. Lasers Med Sci. 2011;26:481-485.
  78. Park KK, Murase JE. Narrowband UV-B phototherapy during pregnancy and folic acid depletion. Arch Dermatol. 2012;148:132-133.
  79. Jablonski NG. A possible link between neural tube defects and ultraviolet light exposure. Med Hypotheses. 1999;52:581-582.
  80. Zhang M, Goyert G, Lim HW. Folate and phototherapy: what should we inform our patients? J Am Acad Dermatol. 2017;77:958-964.
  81. AviClear. Cutera website. Accessed January 8, 2024. https://www.cutera.com/solutions/aviclear/
  82. Wu X, Yang Y, Wang Y, et al. Treatment of refractory acne using selective sebaceous gland electro-thermolysis combined with non-thermal plasma. J Cosmet Laser Ther. 2021;23:188-194.
  83. Ahn GR, Kim JM, Park SJ, et al. Selective sebaceous gland electrothermolysis using a single microneedle radiofrequency device for acne patients: a prospective randomized controlled study. Lasers Surg Med. 2020;52:396-401.
  84. Fabbrocini G, De Padova MP, Tosti A. Chemical peels: what’s new and what isn’t new but still works well. Facial Plast Surg. 2009;25:329-336.
  85. Andersen FA. Final report on the safety assessment of glycolic acid, ammonium, calcium, potassium, and sodium glycolates, methyl, ethyl, propyl, and butyl glycolates, and lactic acid, ammonium, calcium, potassium, sodium, and TEA-lactates, methyl, ethyl, isopropyl, and butyl lactates, and lauryl, myristyl, and cetyl lactates. Int J Toxicol. 1998;17(1_suppl):1-241.
  86. Lee KC, Korgavkar K, Dufresne RG Jr, et al. Safety of cosmetic dermatologic procedures during pregnancy. Dermatol Surg. 2013;39:1573-1586.
  87. James AH, Brancazio LR, Price T. Aspirin and reproductive outcomes. Obstet Gynecol Surv. 2008;63:49-57.
  88. Zhou W-S, Xu L, Xie S-H, et al. Decreased birth weight in relation to maternal urinary trichloroacetic acid levels. Sci Total Environ. 2012;416:105-110.
  89. Schwartz DB, Greenberg MD, Daoud Y, et al. Genital condylomas in pregnancy: use of trichloroacetic acid and laser therapy. Am J Obstet Gynecol. 1988;158:1407-1416.
  90. Starkman SJ, Mangat DS. Chemical peel (deep, medium, light). Facial Plast Surg Clin North Am. 2020;28:45-57.
  91. Trivedi M, Kroumpouzos G, Murase J. A review of the safety of cosmetic procedures during pregnancy and lactation. Int J Womens Dermatol. 2017;3:6-10.
  92. Gallagher T, Taliercio M, Nia JK, et al. Dermatologist use of intralesional triamcinolone in the treatment of acne. J Clin Aesthet Dermatol. 2020;13:41-43.
  93. Zamil DH, Burns EK, Perez-Sanchez A, et al. Risk of birth defects from vitamin A “acne supplements” sold online. Dermatol Pract Concept. 2021;11:e2021075.
Article PDF
Author and Disclosure Information

Drs. Yaghi and Keri are from the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Florida. Dr. Keri also is from Dermatology Service, Miami VA Hospital, Florida. Daniela Baboun is from Herbert Wertheim College of Medicine, Florida International University, Miami.

Dr. Yaghi and Daniela Baboun report no conflict of interest. Dr. Keri is on the advisory board for Ortho Dermatologics, has received research funding from Galderma, and has received honoraria from Merck Manuals.

Correspondence: Jonette E. Keri, MD, PhD, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RSMB Room 2023A, Miami, FL 33136 (jkeri@med.miami.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
E26-E32
Sections
Author and Disclosure Information

Drs. Yaghi and Keri are from the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Florida. Dr. Keri also is from Dermatology Service, Miami VA Hospital, Florida. Daniela Baboun is from Herbert Wertheim College of Medicine, Florida International University, Miami.

Dr. Yaghi and Daniela Baboun report no conflict of interest. Dr. Keri is on the advisory board for Ortho Dermatologics, has received research funding from Galderma, and has received honoraria from Merck Manuals.

Correspondence: Jonette E. Keri, MD, PhD, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RSMB Room 2023A, Miami, FL 33136 (jkeri@med.miami.edu).

Author and Disclosure Information

Drs. Yaghi and Keri are from the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Florida. Dr. Keri also is from Dermatology Service, Miami VA Hospital, Florida. Daniela Baboun is from Herbert Wertheim College of Medicine, Florida International University, Miami.

Dr. Yaghi and Daniela Baboun report no conflict of interest. Dr. Keri is on the advisory board for Ortho Dermatologics, has received research funding from Galderma, and has received honoraria from Merck Manuals.

Correspondence: Jonette E. Keri, MD, PhD, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RSMB Room 2023A, Miami, FL 33136 (jkeri@med.miami.edu).

Article PDF
Article PDF

Acne vulgaris, or acne, is a highly common inflammatory skin disorder affecting up to 85% of the population, and it constitutes the most commonly presenting chief concern in routine dermatology practice.1 Older teenagers and young adults are most often affected by acne.2 Although acne generally is more common in males, adult-onset acne occurs more frequently in women.2,3 Black and Hispanic women are at higher risk for acne compared to those of Asian, White, or Continental Indian descent.4 As such, acne is a common concern in all women of childbearing age.

Concerns for maternal and fetal safety are important therapeutic considerations, especially because hormonal and physiologic changes in pregnancy can lead to onset of inflammatory acne lesions, particularly during the second and third trimesters.5 Female patients younger than 25 years; with a higher body mass index, prior irregular menstruation, or polycystic ovary syndrome; or those experiencing their first pregnancy are thought to be more commonly affected.5-7 In fact, acne affects up to 43% of pregnant women, and lesions typically extend beyond the face to involve the trunk.6,8-10 Importantly, one-third of women with a history of acne experience symptom relapse after disease-free periods, while two-thirds of those with ongoing disease experience symptom deterioration during pregnancy.10 Although acne is not a life-threatening condition, it has a well-documented, detrimental impact on social, emotional, and psychological well-being, namely self-perception, social interactions, quality-of-life scores, depression, and anxiety.11

Therefore, safe and effective treatment of pregnant women is of paramount importance. Because pregnant women are not included in clinical trials, there is a paucity of medication safety data, further augmented by inefficient access to available information. The US Food and Drug Administration (FDA) pregnancy safety categories were updated in 2015, letting go of the traditional A, B, C, D, and X categories.12 The Table reviews the current pregnancy classification system. In this narrative review, we summarize the most recent available data and recommendations on the safety and efficacy of acne treatment during pregnancy.

CT113001026_e_Table.jpg

Topical Treatments for Acne

Benzoyl PeroxideBenzoyl peroxide commonly is used as first-line therapy alone or in combination with other agents for the treatment of mild to moderate acne.13 It is safe for use during pregnancy.14 Although the medication is systemically absorbed, it undergoes complete metabolism to benzoic acid, a commonly used food additive.15,16 Benzoic acid has low bioavailability, as it gets rapidly metabolized by the kidneys; therefore, benzoyl peroxide is unlikely to reach clinically significant levels in the maternal circulation and consequently the fetal circulation. Additionally, it has a low risk for causing congenital malformations.17

Salicylic AcidFor mild to moderate acne, salicylic acid is a second-line agent that likely is safe for use by pregnant women at low concentrations and over limited body surface areas.14,18,19 There is minimal systemic absorption of the drug.20 Additionally, aspirin, which is broken down in the body into salicylic acid, is used in low doses for the treatment of pre-eclampsia during pregnancy.21

DapsoneThe use of dapsone gel 5% as a second-line agent has shown efficacy for mild to moderate acne.22 The oral formulation, commonly used for malaria and leprosy prophylaxis, has failed to show associated fetal toxicity or congenital anomalies.23,24 It also has been used as a first-line treatment for dermatitis herpetiformis in pregnancy.25 Although the medication likely is safe, it is better to minimize its use during the third trimester to reduce the theoretical risk for hyperbilirubinemia in the neonate.17,26-29

Azelaic AcidAzelaic acid effectively targets noninflammatory and inflammatory acne and generally is well tolerated, harboring a good safety profile.30 Topical 20% azelaic acid has localized antibacterial and comedolytic effects and is safe for use during pregnancy.31,32

 

 

Glycolic AcidLimited data exist on the safety of glycolic acid during pregnancy. In vitro studies have shown up to 27% systemic absorption depending on pH, concentration, and duration of application.33 Animal reproductive studies involving rats have shown fetal multisystem malformations and developmental abnormalities with oral administration of glycolic acid at doses far exceeding those used in humans.34 Although no human reproductive studies exist, topical glycolic acid is unlikely to reach the developing fetus in notable amounts, and the medication is likely safe for use.17,35

ClindamycinTopical clindamycin phosphate is an effective and well-tolerated agent for the treatment of mild to moderate acne.36 Its systemic absorption is minimal, and it is considered safe for use during all trimesters of pregnancy.14,17,26,27,35,37

ErythromycinTopical erythromycin is another commonly prescribed topical antibiotic used to target mild to moderate acne. However, its use recently has been associated with a decrease in efficacy secondary to the rise of antibacterial resistance in the community.38-40 Nevertheless, it remains a safe treatment for use during all trimesters of pregnancy.14,17,26,27,35,37

Topical RetinoidsVitamin A derivatives (also known as retinoids) are the mainstay for the treatment of mild to moderate acne. Limited data exist regarding pregnancy outcomes after in utero exposure.41 A rare case report suggested topical tretinoin has been associated with fetal otocerebral anomalies.42 For tazarotene, teratogenic effects were seen in animal reproductive studies at doses exceeding maximum recommended human doses.41,43 However, a large meta-analysis failed to find a clear risk for increased congenital malformations, spontaneous abortions, stillbirth, elective termination of pregnancy, low birthweight, or prematurity following first-trimester exposure to topical retinoids.44 As the level of exposure that could lead to teratogenicity in humans is unknown, avoidance of both tretinoin and tazarotene is recommended in pregnant women.41,45 Nevertheless, women inadvertently exposed should be reassured.44

Conversely, adapalene has been associated with 1 case of anophthalmia and agenesis of the optic chiasma in a fetus following exposure until 13 weeks’ gestation.46 However, a large, open-label trial prior to the patient transitioning from adapalene to over-the-counter treatment showed that the drug harbors a large and reassuring margin of safety and no risk for teratogenicity in a maximal usage trial and Pregnancy Safety Review.47 Therefore, adapalene gel 0.1% is a safe and effective medication for the treatment of acne in a nonprescription environment and does not pose harm to the fetus.

ClascoteroneClascoterone is a novel topical antiandrogenic drug approved for the treatment of hormonal and inflammatory moderate to severe acne.48-51 Human reproductive data are limited to 1 case of pregnancy that occurred during phase 3 trial investigations, and no adverse outcomes were reported.51 Minimal systemic absorption follows topical use.52 Nonetheless, dose-independent malformations were reported in animal reproductive studies.53 As such, it remains better to avoid the use of clascoterone during pregnancy pending further safety data.

Minocycline FoamMinocycline foam 4% is approved to treat inflammatory lesions of nonnodular moderate to severe acne in patients 9 years and older.54 Systemic absorption is minimal, and the drug has limited bioavailability with minimal systemic accumulation in the patient’s serum.55 Given this information, it is unlikely that topical minocycline will reach notable levels in the fetal serum or harbor teratogenic effects, as seen with the oral formulation.56 However, it may be best to avoid its use during the second and third trimesters given the potential risk for tooth discoloration in the fetus.57,58

 

 

Systemic Treatments for Acne

IsotretinoinIsotretinoin is the most effective treatment for moderate to severe acne with a well-documented potential for long-term clearance.59 Its use during pregnancy is absolutely contraindicated, as the medication is a well-known teratogen. Associated congenital malformations include numerous craniofacial defects, cardiovascular and neurologic malformations, or thymic disorders that are estimated to affect 20% to 35% of infants exposed in utero.60 Furthermore, strict contraception use during treatment is mandated for patients who can become pregnant. It is recommended to wait at least 1 month and 1 menstrual cycle after medication discontinuation before attempting to conceive.17 Pregnancy termination is recommended if conception occurs during treatment with isotretinoin.

SpironolactoneSpironolactone is an androgen-receptor antagonist commonly prescribed off label for mild to severe acne in females.61,62 Spironolactone promotes the feminization of male fetuses and should be avoided in pregnancy.63

Doxycycline/MinocyclineTetracyclines are the most commonly prescribed oral antibiotics for moderate to severe acne.64 Although highly effective at treating acne, tetracyclines generally should be avoided in pregnancy. First-trimester use of doxycycline is not absolutely contraindicated but should be reserved for severe illness and not employed for the treatment of acne. However, accidental exposure to doxycycline has not been associated with congenital malformations.65 Nevertheless, after the 15th week of gestation, permanent tooth discoloration and bone growth inhibition in the fetus are serious and well-documented risks.14,17 Additional adverse events following in utero exposure include infantile inguinal hernia, hypospadias, and limb hypoplasia.63

SarecyclineSarecycline is a novel tetracycline-class antibiotic for the treatment of moderate to severe inflammatory acne. It has a narrower spectrum of activity compared to its counterparts within its class, which translates to an improved safety profile, namely when it comes to gastrointestinal tract microbiome disruption and potentially decreased likelihood of developing bacterial resistance.66 Data on human reproductive studies are limited, but it is advisable to avoid sarecycline in pregnancy, as it may cause adverse developmental effects in the fetus, such as reduced bone growth, in addition to the well-known tetracycline-associated risk for permanent discoloration of the teeth if used during the second and third trimesters.67,68

ErythromycinOral erythromycin targets moderate to severe inflammatory acne and is considered safe for use during pregnancy.69,70 There has been 1 study reporting an increased risk for atrial and ventricular septal defects (1.8%) and pyloric stenosis (0.2%), but these risks are still uncertain, and erythromycin is considered compatible with pregnancy.71 However, erythromycin estolate formulations should be avoided given the associated 10% to 15% risk for reversible cholestatic liver injury.72 Erythromycin base or erythromycin ethylsuccinate formulations should be favored.

Systemic SteroidsPrednisone is indicated for severe acne with scarring and should only be used during pregnancy after clearance from the patient’s obstetrician. Doses of 0.5 mg/kg or less should be prescribed in combination with systemic antibiotics as well as agents for bone and gastrointestinal tract prophylaxis.29

ZincThe exact mechanism by which zinc exerts its effects to improve acne remains largely obscure. It has been found effective against inflammatory lesions of mild to moderate acne.73 Generally recommended dosages range from 30 to 200 mg/d but may be associated with gastrointestinal tract disturbances. Dosages of 75 mg/d have shown no harm to the fetus.74 When taking this supplement, patients should not exceed the recommended doses given the risk for hypocupremia associated with high-dose zinc supplementation.

 

 

Light-Based Therapies

PhototherapyNarrowband UVB phototherapy is effective for the treatment of mild to moderate acne.75 It has been proven to be a safe treatment option during pregnancy, but its use has been associated with decreased folic acid levels.76-79 Therefore, in addition to attaining baseline folic acid serum levels, supplementation with folic acid prior to treatment, as per routine prenatal guidelines, should be sought.80

AviClearThe AviClear (Cutera) laser is the first device cleared by the FDA for mild to severe acne in March 2022.81 The FDA clearance for the Accure (Accure Acne Inc) laser, also targeting mild to severe acne, followed soon after (November 2022). Both lasers harbor a wavelength of 1726 nm and target sebaceous glands with electrothermolysis.82,83 Further research and long-term safety data are required before using them in pregnancy.

Other Therapies

Cosmetic PeelsGlycolic acid peels induce epidermolysis and desquamation.84 Although data on use during pregnancy are limited, these peels have limited dermal penetration and are considered safe for use in pregnancy.33,85,86 Similarly, keratolytic lactic acid peels harbor limited dermal penetration and can be safely used in pregnant women.87-89 Salicylic acid peels also work through epidermolysis and desquamation84; however, they tend to penetrate deeper into the skin, reaching down to the basal layer, if large areas are treated or when applied under occlusion.86,90 Although their use is not contraindicated in pregnancy, they should be limited to small areas of coverage.91

Intralesional TriamcinoloneAcne cysts and inflammatory papules can be treated with intralesional triamcinolone injections to relieve acute symptoms such as pain.92 Low doses at concentrations of 2.5 mg/mL are considered compatible with pregnancy when indicated.29

Approaching the Patient Clinical Encounter

In patients seeking treatment prior to conception, a few recommendations can be made to minimize the risk for acne recurrence or flares during pregnancy. For instance, because data show an association between increased acne severity in those with a higher body mass index and in pregnancy, weight loss may be recommended prior to pregnancy to help mitigate symptoms after conception.7 The Figure summarizes our recommendations for approaching and treating acne in pregnancy.

ret_Yaghi_figure.jpg
%3Cp%3EAn%20algorithm-based%20approach%20for%20the%20management%20of%20acne%20during%20pregnancy.%3C%2Fp%3E

In all patients, grading the severity of the patient’s acne as mild, moderate, or severe is the first step. The presence of scarring is an additional consideration during the physical examination and should be documented. A careful discussion of treatment expectations and prognosis should be the focus before treatment initiation. Meticulous documentation of the physical examination and discussion with the patient should be prioritized.

To minimize toxicity and risks to the developing fetus, monotherapy is favored. Topical therapy should be considered first line. Safe regimens include mild nonabrasive washes, such as those containing benzoyl peroxide or glycolic acid, or topical azelaic acid or clindamycin phosphate for mild to moderate acne. More severe cases warrant the consideration of systemic medications as second line, as more severe acne is better treated with oral antibiotics such as the macrolides erythromycin or clindamycin or systemic corticosteroids when concern exists for severe scarring. The additional use of physical sunscreen also is recommended.

An important topic to address during the clinical encounter is cautious intake of oral supplements for acne during pregnancy, as they may contain harmful and teratogenic ingredients. A recent search focusing on acne supplements available online between March and May 2020 uncovered 49 different supplements, 26 (53%) of which contained vitamin A.93 Importantly, 3 (6%) of these 49 supplements were likely teratogenic, 4 (8%) contained vitamin A doses exceeding the recommended daily nutritional intake level, and 15 (31%) harbored an unknown teratogenic risk. Furthermore, among the 6 (12%) supplements with vitamin A levels exceeding 10,000 IU, 2 lacked any mention of pregnancy warning, including the supplement with the highest vitamin A dose found in this study.93 Because dietary supplements are not subject to the same stringent regulations by the FDA as drugs, inadvertent use by unaware patients ought to be prevented by careful counseling and education.

Finally, patients should be counseled to seek care following delivery for potentially updated medication management of acne, especially if they are breastfeeding. Co-management with a pediatrician may be indicated during lactation, particularly when newborns are born preterm or with other health conditions that may warrant additional caution with the use of certain agents.

Acne vulgaris, or acne, is a highly common inflammatory skin disorder affecting up to 85% of the population, and it constitutes the most commonly presenting chief concern in routine dermatology practice.1 Older teenagers and young adults are most often affected by acne.2 Although acne generally is more common in males, adult-onset acne occurs more frequently in women.2,3 Black and Hispanic women are at higher risk for acne compared to those of Asian, White, or Continental Indian descent.4 As such, acne is a common concern in all women of childbearing age.

Concerns for maternal and fetal safety are important therapeutic considerations, especially because hormonal and physiologic changes in pregnancy can lead to onset of inflammatory acne lesions, particularly during the second and third trimesters.5 Female patients younger than 25 years; with a higher body mass index, prior irregular menstruation, or polycystic ovary syndrome; or those experiencing their first pregnancy are thought to be more commonly affected.5-7 In fact, acne affects up to 43% of pregnant women, and lesions typically extend beyond the face to involve the trunk.6,8-10 Importantly, one-third of women with a history of acne experience symptom relapse after disease-free periods, while two-thirds of those with ongoing disease experience symptom deterioration during pregnancy.10 Although acne is not a life-threatening condition, it has a well-documented, detrimental impact on social, emotional, and psychological well-being, namely self-perception, social interactions, quality-of-life scores, depression, and anxiety.11

Therefore, safe and effective treatment of pregnant women is of paramount importance. Because pregnant women are not included in clinical trials, there is a paucity of medication safety data, further augmented by inefficient access to available information. The US Food and Drug Administration (FDA) pregnancy safety categories were updated in 2015, letting go of the traditional A, B, C, D, and X categories.12 The Table reviews the current pregnancy classification system. In this narrative review, we summarize the most recent available data and recommendations on the safety and efficacy of acne treatment during pregnancy.

CT113001026_e_Table.jpg

Topical Treatments for Acne

Benzoyl PeroxideBenzoyl peroxide commonly is used as first-line therapy alone or in combination with other agents for the treatment of mild to moderate acne.13 It is safe for use during pregnancy.14 Although the medication is systemically absorbed, it undergoes complete metabolism to benzoic acid, a commonly used food additive.15,16 Benzoic acid has low bioavailability, as it gets rapidly metabolized by the kidneys; therefore, benzoyl peroxide is unlikely to reach clinically significant levels in the maternal circulation and consequently the fetal circulation. Additionally, it has a low risk for causing congenital malformations.17

Salicylic AcidFor mild to moderate acne, salicylic acid is a second-line agent that likely is safe for use by pregnant women at low concentrations and over limited body surface areas.14,18,19 There is minimal systemic absorption of the drug.20 Additionally, aspirin, which is broken down in the body into salicylic acid, is used in low doses for the treatment of pre-eclampsia during pregnancy.21

DapsoneThe use of dapsone gel 5% as a second-line agent has shown efficacy for mild to moderate acne.22 The oral formulation, commonly used for malaria and leprosy prophylaxis, has failed to show associated fetal toxicity or congenital anomalies.23,24 It also has been used as a first-line treatment for dermatitis herpetiformis in pregnancy.25 Although the medication likely is safe, it is better to minimize its use during the third trimester to reduce the theoretical risk for hyperbilirubinemia in the neonate.17,26-29

Azelaic AcidAzelaic acid effectively targets noninflammatory and inflammatory acne and generally is well tolerated, harboring a good safety profile.30 Topical 20% azelaic acid has localized antibacterial and comedolytic effects and is safe for use during pregnancy.31,32

 

 

Glycolic AcidLimited data exist on the safety of glycolic acid during pregnancy. In vitro studies have shown up to 27% systemic absorption depending on pH, concentration, and duration of application.33 Animal reproductive studies involving rats have shown fetal multisystem malformations and developmental abnormalities with oral administration of glycolic acid at doses far exceeding those used in humans.34 Although no human reproductive studies exist, topical glycolic acid is unlikely to reach the developing fetus in notable amounts, and the medication is likely safe for use.17,35

ClindamycinTopical clindamycin phosphate is an effective and well-tolerated agent for the treatment of mild to moderate acne.36 Its systemic absorption is minimal, and it is considered safe for use during all trimesters of pregnancy.14,17,26,27,35,37

ErythromycinTopical erythromycin is another commonly prescribed topical antibiotic used to target mild to moderate acne. However, its use recently has been associated with a decrease in efficacy secondary to the rise of antibacterial resistance in the community.38-40 Nevertheless, it remains a safe treatment for use during all trimesters of pregnancy.14,17,26,27,35,37

Topical RetinoidsVitamin A derivatives (also known as retinoids) are the mainstay for the treatment of mild to moderate acne. Limited data exist regarding pregnancy outcomes after in utero exposure.41 A rare case report suggested topical tretinoin has been associated with fetal otocerebral anomalies.42 For tazarotene, teratogenic effects were seen in animal reproductive studies at doses exceeding maximum recommended human doses.41,43 However, a large meta-analysis failed to find a clear risk for increased congenital malformations, spontaneous abortions, stillbirth, elective termination of pregnancy, low birthweight, or prematurity following first-trimester exposure to topical retinoids.44 As the level of exposure that could lead to teratogenicity in humans is unknown, avoidance of both tretinoin and tazarotene is recommended in pregnant women.41,45 Nevertheless, women inadvertently exposed should be reassured.44

Conversely, adapalene has been associated with 1 case of anophthalmia and agenesis of the optic chiasma in a fetus following exposure until 13 weeks’ gestation.46 However, a large, open-label trial prior to the patient transitioning from adapalene to over-the-counter treatment showed that the drug harbors a large and reassuring margin of safety and no risk for teratogenicity in a maximal usage trial and Pregnancy Safety Review.47 Therefore, adapalene gel 0.1% is a safe and effective medication for the treatment of acne in a nonprescription environment and does not pose harm to the fetus.

ClascoteroneClascoterone is a novel topical antiandrogenic drug approved for the treatment of hormonal and inflammatory moderate to severe acne.48-51 Human reproductive data are limited to 1 case of pregnancy that occurred during phase 3 trial investigations, and no adverse outcomes were reported.51 Minimal systemic absorption follows topical use.52 Nonetheless, dose-independent malformations were reported in animal reproductive studies.53 As such, it remains better to avoid the use of clascoterone during pregnancy pending further safety data.

Minocycline FoamMinocycline foam 4% is approved to treat inflammatory lesions of nonnodular moderate to severe acne in patients 9 years and older.54 Systemic absorption is minimal, and the drug has limited bioavailability with minimal systemic accumulation in the patient’s serum.55 Given this information, it is unlikely that topical minocycline will reach notable levels in the fetal serum or harbor teratogenic effects, as seen with the oral formulation.56 However, it may be best to avoid its use during the second and third trimesters given the potential risk for tooth discoloration in the fetus.57,58

 

 

Systemic Treatments for Acne

IsotretinoinIsotretinoin is the most effective treatment for moderate to severe acne with a well-documented potential for long-term clearance.59 Its use during pregnancy is absolutely contraindicated, as the medication is a well-known teratogen. Associated congenital malformations include numerous craniofacial defects, cardiovascular and neurologic malformations, or thymic disorders that are estimated to affect 20% to 35% of infants exposed in utero.60 Furthermore, strict contraception use during treatment is mandated for patients who can become pregnant. It is recommended to wait at least 1 month and 1 menstrual cycle after medication discontinuation before attempting to conceive.17 Pregnancy termination is recommended if conception occurs during treatment with isotretinoin.

SpironolactoneSpironolactone is an androgen-receptor antagonist commonly prescribed off label for mild to severe acne in females.61,62 Spironolactone promotes the feminization of male fetuses and should be avoided in pregnancy.63

Doxycycline/MinocyclineTetracyclines are the most commonly prescribed oral antibiotics for moderate to severe acne.64 Although highly effective at treating acne, tetracyclines generally should be avoided in pregnancy. First-trimester use of doxycycline is not absolutely contraindicated but should be reserved for severe illness and not employed for the treatment of acne. However, accidental exposure to doxycycline has not been associated with congenital malformations.65 Nevertheless, after the 15th week of gestation, permanent tooth discoloration and bone growth inhibition in the fetus are serious and well-documented risks.14,17 Additional adverse events following in utero exposure include infantile inguinal hernia, hypospadias, and limb hypoplasia.63

SarecyclineSarecycline is a novel tetracycline-class antibiotic for the treatment of moderate to severe inflammatory acne. It has a narrower spectrum of activity compared to its counterparts within its class, which translates to an improved safety profile, namely when it comes to gastrointestinal tract microbiome disruption and potentially decreased likelihood of developing bacterial resistance.66 Data on human reproductive studies are limited, but it is advisable to avoid sarecycline in pregnancy, as it may cause adverse developmental effects in the fetus, such as reduced bone growth, in addition to the well-known tetracycline-associated risk for permanent discoloration of the teeth if used during the second and third trimesters.67,68

ErythromycinOral erythromycin targets moderate to severe inflammatory acne and is considered safe for use during pregnancy.69,70 There has been 1 study reporting an increased risk for atrial and ventricular septal defects (1.8%) and pyloric stenosis (0.2%), but these risks are still uncertain, and erythromycin is considered compatible with pregnancy.71 However, erythromycin estolate formulations should be avoided given the associated 10% to 15% risk for reversible cholestatic liver injury.72 Erythromycin base or erythromycin ethylsuccinate formulations should be favored.

Systemic SteroidsPrednisone is indicated for severe acne with scarring and should only be used during pregnancy after clearance from the patient’s obstetrician. Doses of 0.5 mg/kg or less should be prescribed in combination with systemic antibiotics as well as agents for bone and gastrointestinal tract prophylaxis.29

ZincThe exact mechanism by which zinc exerts its effects to improve acne remains largely obscure. It has been found effective against inflammatory lesions of mild to moderate acne.73 Generally recommended dosages range from 30 to 200 mg/d but may be associated with gastrointestinal tract disturbances. Dosages of 75 mg/d have shown no harm to the fetus.74 When taking this supplement, patients should not exceed the recommended doses given the risk for hypocupremia associated with high-dose zinc supplementation.

 

 

Light-Based Therapies

PhototherapyNarrowband UVB phototherapy is effective for the treatment of mild to moderate acne.75 It has been proven to be a safe treatment option during pregnancy, but its use has been associated with decreased folic acid levels.76-79 Therefore, in addition to attaining baseline folic acid serum levels, supplementation with folic acid prior to treatment, as per routine prenatal guidelines, should be sought.80

AviClearThe AviClear (Cutera) laser is the first device cleared by the FDA for mild to severe acne in March 2022.81 The FDA clearance for the Accure (Accure Acne Inc) laser, also targeting mild to severe acne, followed soon after (November 2022). Both lasers harbor a wavelength of 1726 nm and target sebaceous glands with electrothermolysis.82,83 Further research and long-term safety data are required before using them in pregnancy.

Other Therapies

Cosmetic PeelsGlycolic acid peels induce epidermolysis and desquamation.84 Although data on use during pregnancy are limited, these peels have limited dermal penetration and are considered safe for use in pregnancy.33,85,86 Similarly, keratolytic lactic acid peels harbor limited dermal penetration and can be safely used in pregnant women.87-89 Salicylic acid peels also work through epidermolysis and desquamation84; however, they tend to penetrate deeper into the skin, reaching down to the basal layer, if large areas are treated or when applied under occlusion.86,90 Although their use is not contraindicated in pregnancy, they should be limited to small areas of coverage.91

Intralesional TriamcinoloneAcne cysts and inflammatory papules can be treated with intralesional triamcinolone injections to relieve acute symptoms such as pain.92 Low doses at concentrations of 2.5 mg/mL are considered compatible with pregnancy when indicated.29

Approaching the Patient Clinical Encounter

In patients seeking treatment prior to conception, a few recommendations can be made to minimize the risk for acne recurrence or flares during pregnancy. For instance, because data show an association between increased acne severity in those with a higher body mass index and in pregnancy, weight loss may be recommended prior to pregnancy to help mitigate symptoms after conception.7 The Figure summarizes our recommendations for approaching and treating acne in pregnancy.

ret_Yaghi_figure.jpg
%3Cp%3EAn%20algorithm-based%20approach%20for%20the%20management%20of%20acne%20during%20pregnancy.%3C%2Fp%3E

In all patients, grading the severity of the patient’s acne as mild, moderate, or severe is the first step. The presence of scarring is an additional consideration during the physical examination and should be documented. A careful discussion of treatment expectations and prognosis should be the focus before treatment initiation. Meticulous documentation of the physical examination and discussion with the patient should be prioritized.

To minimize toxicity and risks to the developing fetus, monotherapy is favored. Topical therapy should be considered first line. Safe regimens include mild nonabrasive washes, such as those containing benzoyl peroxide or glycolic acid, or topical azelaic acid or clindamycin phosphate for mild to moderate acne. More severe cases warrant the consideration of systemic medications as second line, as more severe acne is better treated with oral antibiotics such as the macrolides erythromycin or clindamycin or systemic corticosteroids when concern exists for severe scarring. The additional use of physical sunscreen also is recommended.

An important topic to address during the clinical encounter is cautious intake of oral supplements for acne during pregnancy, as they may contain harmful and teratogenic ingredients. A recent search focusing on acne supplements available online between March and May 2020 uncovered 49 different supplements, 26 (53%) of which contained vitamin A.93 Importantly, 3 (6%) of these 49 supplements were likely teratogenic, 4 (8%) contained vitamin A doses exceeding the recommended daily nutritional intake level, and 15 (31%) harbored an unknown teratogenic risk. Furthermore, among the 6 (12%) supplements with vitamin A levels exceeding 10,000 IU, 2 lacked any mention of pregnancy warning, including the supplement with the highest vitamin A dose found in this study.93 Because dietary supplements are not subject to the same stringent regulations by the FDA as drugs, inadvertent use by unaware patients ought to be prevented by careful counseling and education.

Finally, patients should be counseled to seek care following delivery for potentially updated medication management of acne, especially if they are breastfeeding. Co-management with a pediatrician may be indicated during lactation, particularly when newborns are born preterm or with other health conditions that may warrant additional caution with the use of certain agents.

References
  1. Bhate K, Williams H. Epidemiology of acne vulgaris. Br J Dermatol. 2013;168:474-485.
  2. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020;10:5754.
  3. Fisk WA, Lev-Tov HA, Sivamani RK. Epidemiology and management of acne in adult women. Curr Dermatol Rep. 2014;3:29-39.
  4. Perkins A, Cheng C, Hillebrand G, et al. Comparison of the epidemiology of acne vulgaris among Caucasian, Asian, Continental Indian and African American women. J Eur Acad Dermatol Venereol. 2011;25:1054-1060.
  5. Yang CC, Huang YT, Yu CH, et al. Inflammatory facial acne during uncomplicated pregnancy and post‐partum in adult women: a preliminary hospital‐based prospective observational study of 35 cases from Taiwan. J Eur Acad Dermatol Venereol. 2016;30:1787-1789.
  6. Dréno B, Blouin E, Moyse D, et al. Acne in pregnant women: a French survey. Acta Derm Venereol. 2014;94:82-83.
  7. Kutlu Ö, Karadag˘ AS, Ünal E, et al. Acne in pregnancy: a prospective multicenter, cross‐sectional study of 295 patients in Turkey. Int J Dermatol. 2020;59:1098-1105.
  8. Hoefel IDR, Weber MB, Manzoni APD, et al. Striae gravidarum, acne, facial spots, and hair disorders: risk factors in a study with 1284 puerperal patients. J Pregnancy. 2020;2020:8036109.
  9. Ayanlowo OO, Otrofanowei E, Shorunmu TO, et al. Pregnancy dermatoses: a study of patients attending the antenatal clinic at two tertiary care centers in south west Nigeria. PAMJ Clin Med. 2020;3.
  10. Bechstein S, Ochsendorf F. Acne and rosacea in pregnancy. Hautarzt. 2017;68:111-119.
  11. Habeshian KA, Cohen BA. Current issues in the treatment of acne vulgaris. Pediatrics. 2020;145(suppl 2):S225-S230.
  12. Content and format of labeling for human prescription drug and biological products; requirements for pregnancy and lactation labeling (21 CFR 201). Fed Regist. 2014;79:72064-72103.
  13. Sagransky M, Yentzer BA, Feldman SR. Benzoyl peroxide: a review of its current use in the treatment of acne vulgaris. Expert Opin Pharmacother. 2009;10:2555-2562.
  14. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation: part I. Pregnancy. J Am Acad Dermatol. 2014;70:401.e1-401.e14; quiz 415.
  15. Wolverton SE. Systemic corticosteroids. Comprehensive Dermatol Drug Ther. 2012;3:143-168.
  16. Kirtschig G, Schaefer C. Dermatological medications and local therapeutics. In: Schaefer C, Peters P, Miller RK, eds. Drugs During Pregnancy and Lactation. 3rd edition. Elsevier; 2015:467-492.
  17. Pugashetti R, Shinkai K. Treatment of acne vulgaris in pregnant patients. Dermatol Ther. 2013;26:302-311.
  18. Touitou E, Godin B, Shumilov M, et al. Efficacy and tolerability of clindamycin phosphate and salicylic acid gel in the treatment of mild to moderate acne vulgaris. J Eur Acad Dermatol Venereol. 2008;22:629-631.
  19. Schaefer C, Peters PW, Miller RK, eds. Drugs During Pregnancy and Lactation: Treatment Options and Risk Assessment. 2nd ed. Academic Press; 2014.
  20. Birmingham B, Greene D, Rhodes C. Systemic absorption of topical salicylic acid. Int J Dermatol. 1979;18:228-231.
  21. Trivedi NA. A meta-analysis of low-dose aspirin for prevention of preeclampsia. J Postgrad Med. 2011;57:91-95.
  22. Lucky AW, Maloney JM, Roberts J, et al. Dapsone gel 5% for the treatment of acne vulgaris: safety and efficacy of long-term (1 year) treatment. J Drugs Dermatol. 2007;6:981-987.
  23. Nosten F, McGready R, d’Alessandro U, et al. Antimalarial drugs in pregnancy: a review. Curr Drug Saf. 2006;1:1-15.
  24. Brabin BJ, Eggelte TA, Parise M, et al. Dapsone therapy for malaria during pregnancy: maternal and fetal outcomes. Drug Saf. 2004;27:633-648.
  25. Tuffanelli DL. Successful pregnancy in a patient with dermatitis herpetiformis treated with low-dose dapsone. Arch Dermatol. 1982;118:876.
  26. Meredith FM, Ormerod AD. The management of acne vulgaris in pregnancy. Am J Clin Dermatol. 2013;14:351-358.
  27. Kong Y, Tey H. Treatment of acne vulgaris during pregnancy and lactation. Drugs. 2013;73:779-787.
  28. Leachman SA, Reed BR. The use of dermatologic drugs in pregnancy and lactation. Dermatol Clin. 2006;24:167-197.
  29. Ly S, Kamal K, Manjaly P, et al. Treatment of acne vulgaris during pregnancy and lactation: a narrative review. Dermatol Ther. 2023;13:115-130.
  30. Webster G. Combination azelaic acid therapy for acne vulgaris. J Am Acad Dermatol. 2000;43:S47-S50.
  31. Archer CB, Cohen SN, Baron SE. Guidance on the diagnosis and clinical management of acne. Clin Exp Dermatol. 2012;37(suppl 1):1-6.
  32. Graupe K, Cunliffe W, Gollnick H, et al. Efficacy and safety of topical azelaic acid (20 percent cream): an overview of results from European clinical trials and experimental reports. Cutis. 1996;57(1 suppl):20-35.
  33. Bozzo P, Chua-Gocheco A, Einarson A. Safety of skin care products during pregnancy. Can Fam Physician. 2011;57:665-667.
  34. Munley SM, Kennedy GL, Hurtt ME. Developmental toxicity study of glycolic acid in rats. Drug Chem Toxicol. 1999;22:569-582.
  35. Chien AL, Qi J, Rainer B, et al. Treatment of acne in pregnancy. J Am Board Fam Med. 2016;29:254-262.
  36. Stuart B, Maund E, Wilcox C, et al. Topical preparations for the treatment of mild‐to‐moderate acne vulgaris: systematic review and network meta‐analysis. Br J Dermatol. 2021;185:512-525.
  37. van Hoogdalem EJ, Baven TL, Spiegel‐Melsen I, et al. Transdermal absorption of clindamycin and tretinoin from topically applied anti‐acne formulations in man. Biopharm Drug Dispos. 1998;19:563-569.
  38. Austin BA, Fleischer AB Jr. The extinction of topical erythromycin therapy for acne vulgaris and concern for the future of topical clindamycin. J Dermatolog Treat. 2017;28:145-148.
  39. Eady EA, Cove J, Holland K, et al. Erythromycin resistant propionibacteria in antibiotic treated acne patients: association with therapeutic failure. Br J. Dermatol. 1989;121:51-57.
  40. Alkhawaja E, Hammadi S, Abdelmalek M, et al. Antibiotic resistant Cutibacterium acnes among acne patients in Jordan: a cross sectional study. BMC Dermatol. 2020;20:1-9.
  41. Han G, Wu JJ, Del Rosso JQ. Use of topical tazarotene for the treatment of acne vulgaris in pregnancy: a literature review. J Clin Aesthet Dermatol. 2020;13:E59-E65.
  42. Selcen D, Seidman S, Nigro MA. Otocerebral anomalies associated with topical tretinoin use. Brain Dev. 2000;22:218-220.
  43. Moretz D. Drug Class Update with New Drug Evaluations: Topical Products for Inflammatory Skin Conditions. Oregon State University Drug Use & Research Management Program; December 2022. Accessed January 8, 2024. https://www.orpdl.org/durm/meetings/meetingdocs/2022_12_01/archives/2022_12_01_Inflammatory_Skin_Dz_ClassUpdate.pdf
  44. Kaplan YC, Ozsarfati J, Etwel F, et al. Pregnancy outcomes following first‐trimester exposure to topical retinoids: a systematic review and meta‐analysis. Br J Dermatol. 2015;173:1132-1141.
  45. Menter A. Pharmacokinetics and safety of tazarotene. J Am Acad Dermatol. 2000;43(2, pt 3):S31-S35.
  46. Autret E, Berjot M, Jonville-Béra A-P, et al. Anophthalmia and agenesis of optic chiasma associated with adapalene gel in early pregnancy. Lancet. 1997;350:339.
  47. Weiss J, Mallavalli S, Meckfessel M, et al. Safe use of adapalene 0.1% gel in a non-prescription environment. J Drugs Dermatol. 2021;20:1330-1335.
  48. Alessandro Mazzetti M. A phase 2b, randomized, double-blind vehicle controlled, dose escalation study evaluating clascoterone 0.1%, 0.5%, and 1% topical cream in subjects with facial acne. J Drugs Dermatol. 2019;18:570-575.
  49. Eichenfield L, Hebert A, Gold LS, et al. Open-label, long-term extension study to evaluate the safety of clascoterone (CB-03-01) cream, 1% twice daily, in patients with acne vulgaris. J Am Acad Dermatol. 2020;83:477-485.
  50. Trifu V, Tiplica GS, Naumescu E, et al. Cortexolone 17α‐propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. a pilot randomized, double‐blind comparative study vs. placebo and tretinoin 0.05% cream. Br J Dermatol. 2011;165:177-183.
  51. Hebert A, Thiboutot D, Gold LS, et al. Efficacy and safety of topical clascoterone cream, 1%, for treatment in patients with facial acne: two phase 3 randomized clinical trials. JAMA Dermatol. 2020;156:621-630.
  52. Alkhodaidi ST, Al Hawsawi KA, Alkhudaidi IT, et al. Efficacy and safety of topical clascoterone cream for treatment of acne vulgaris: a systematic review and meta‐analysis of randomized placebo‐controlled trials. Dermatol Ther. 2021;34:e14609.
  53. Clasoterone. Package insert. Cassiopea Inc; 2020.
  54. Paik J. Topical minocycline foam 4%: a review in acne vulgaris. Am J Clin Dermatol. 2020;21:449-456.
  55. Jones TM, Ellman H. Pharmacokinetic comparison of once-daily topical minocycline foam 4% vs oral minocycline for moderate-to-severe acne. J Drugs Dermatol. 2017;16:1022-1028.
  56. Minocycline hydrochloride extended-release tablets. Package insert. JG Pharma; July 2020. Accessed January 8, 2024. https://www.jgpharmainc.com/assets/pdf/minocycline-hydrochloride.pdf
  57. Dinnendahl V, Fricke U (eds). Arzneistoff-Profile: Basisinformation über arzneiliche Wirkstoffe. Govi Pharmazeutischer Verlag; 2010.
  58. Martins AM, Marto JM, Johnson JL, et al. A review of systemic minocycline side effects and topical minocycline as a safer alternative for treating acne and rosacea. Antibiotics. 2021;10:757.
  59. Landis MN. Optimizing isotretinoin treatment of acne: update on current recommendations for monitoring, dosing, safety, adverse effects, compliance, and outcomes. Am J Clin Dermatol. 2020;21:411-419.
  60. Draghici C-C, Miulescu R-G, Petca R-C, et al. Teratogenic effect of isotretinoin in both fertile females and males. Exp Ther Med. 2021;21:1-5.
  61. Barker RA, Wilcox C, Layton AM. Oral spironolactone for acne vulgaris in adult females: an update of the literature. Am J Clin Dermatol. 2020;21:303-305.
  62. Han JJ, Faletsky A, Barbieri JS, et al. New acne therapies and updates on use of spironolactone and isotretinoin: a narrative review. Dermatol Ther (Heidelb). 2021;11:79-91.
  63. Briggs GG, Freeman RK, Yaffe SJ. Drugs in Pregnancy and Lactation: A Reference Guide to Fetal and Neonatal Risk. Lippincott Williams & Wilkins; 2012.
  64. Patel DJ, Bhatia N. Oral antibiotics for acne. Am J Clin Dermatol. 2021;22:193-204.
  65. Jick H, Holmes LB, Hunter JR, et al. First-trimester drug use and congenital disorders. JAMA. 1981;246:343-346.
  66. Valente Duarte de Sousa IC. An overview of sarecycline for the treatment of moderate-to-severe acne vulgaris. Exp Opin Pharmacother. 2021;22:145-154.
  67. Hussar DA, Chahine EB. Omadacycline tosylate, sarecycline hydrochloride, rifamycin sodium, and moxidectin. J Am Pharm Assoc. 2019;59:756-760.
  68. Haidari W, Bruinsma R, Cardenas-de la Garza JA, et al. Sarecycline review. Ann Pharmacother. 2020;54:164-170.
  69. Feldman S, Careccia RE, Barham KL, et al. Diagnosis and treatment of acne. Am Fam Physician. 2004;69:2123-2130.
  70. Gammon WR, Meyer C, Lantis S, et al. Comparative efficacy of oral erythromycin versus oral tetracycline in the treatment of acne vulgaris: a double-blind study. J Am Acad Dermatol. 1986;14:183-186.
  71. Källén BA, Olausson PO, Danielsson BR. Is erythromycin therapy teratogenic in humans? Reprod Toxicol. 2005;20:209-214.
  72. McCormack WM, George H, Donner A, et al. Hepatotoxicity of erythromycin estolate during pregnancy. Antimicrob Agents Chemother. 1977;12:630-635.
  73. Cervantes J, Eber AE, Perper M, et al. The role of zinc in the treatment of acne: a review of the literature. Dermatolog Ther. 2018;31:e12576.
  74. Dréno B, Blouin E. Acne, pregnant women and zinc salts: a literature review [in French]. Ann Dermatol Venereol. 2008;135:27-33.
  75. Eid MM, Saleh MS, Allam NM, et al. Narrow band ultraviolet B versus red light-emitting diodes in the treatment of facial acne vulgaris: a randomized controlled trial. Photobiomodul Photomed Laser Surg. 2021;39:418-424.
  76. Zeichner JA. Narrowband UV-B phototherapy for the treatment of acne vulgaris during pregnancy. Arch Dermatol. 2011;147:537-539.
  77. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. Lasers Med Sci. 2011;26:481-485.
  78. Park KK, Murase JE. Narrowband UV-B phototherapy during pregnancy and folic acid depletion. Arch Dermatol. 2012;148:132-133.
  79. Jablonski NG. A possible link between neural tube defects and ultraviolet light exposure. Med Hypotheses. 1999;52:581-582.
  80. Zhang M, Goyert G, Lim HW. Folate and phototherapy: what should we inform our patients? J Am Acad Dermatol. 2017;77:958-964.
  81. AviClear. Cutera website. Accessed January 8, 2024. https://www.cutera.com/solutions/aviclear/
  82. Wu X, Yang Y, Wang Y, et al. Treatment of refractory acne using selective sebaceous gland electro-thermolysis combined with non-thermal plasma. J Cosmet Laser Ther. 2021;23:188-194.
  83. Ahn GR, Kim JM, Park SJ, et al. Selective sebaceous gland electrothermolysis using a single microneedle radiofrequency device for acne patients: a prospective randomized controlled study. Lasers Surg Med. 2020;52:396-401.
  84. Fabbrocini G, De Padova MP, Tosti A. Chemical peels: what’s new and what isn’t new but still works well. Facial Plast Surg. 2009;25:329-336.
  85. Andersen FA. Final report on the safety assessment of glycolic acid, ammonium, calcium, potassium, and sodium glycolates, methyl, ethyl, propyl, and butyl glycolates, and lactic acid, ammonium, calcium, potassium, sodium, and TEA-lactates, methyl, ethyl, isopropyl, and butyl lactates, and lauryl, myristyl, and cetyl lactates. Int J Toxicol. 1998;17(1_suppl):1-241.
  86. Lee KC, Korgavkar K, Dufresne RG Jr, et al. Safety of cosmetic dermatologic procedures during pregnancy. Dermatol Surg. 2013;39:1573-1586.
  87. James AH, Brancazio LR, Price T. Aspirin and reproductive outcomes. Obstet Gynecol Surv. 2008;63:49-57.
  88. Zhou W-S, Xu L, Xie S-H, et al. Decreased birth weight in relation to maternal urinary trichloroacetic acid levels. Sci Total Environ. 2012;416:105-110.
  89. Schwartz DB, Greenberg MD, Daoud Y, et al. Genital condylomas in pregnancy: use of trichloroacetic acid and laser therapy. Am J Obstet Gynecol. 1988;158:1407-1416.
  90. Starkman SJ, Mangat DS. Chemical peel (deep, medium, light). Facial Plast Surg Clin North Am. 2020;28:45-57.
  91. Trivedi M, Kroumpouzos G, Murase J. A review of the safety of cosmetic procedures during pregnancy and lactation. Int J Womens Dermatol. 2017;3:6-10.
  92. Gallagher T, Taliercio M, Nia JK, et al. Dermatologist use of intralesional triamcinolone in the treatment of acne. J Clin Aesthet Dermatol. 2020;13:41-43.
  93. Zamil DH, Burns EK, Perez-Sanchez A, et al. Risk of birth defects from vitamin A “acne supplements” sold online. Dermatol Pract Concept. 2021;11:e2021075.
References
  1. Bhate K, Williams H. Epidemiology of acne vulgaris. Br J Dermatol. 2013;168:474-485.
  2. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020;10:5754.
  3. Fisk WA, Lev-Tov HA, Sivamani RK. Epidemiology and management of acne in adult women. Curr Dermatol Rep. 2014;3:29-39.
  4. Perkins A, Cheng C, Hillebrand G, et al. Comparison of the epidemiology of acne vulgaris among Caucasian, Asian, Continental Indian and African American women. J Eur Acad Dermatol Venereol. 2011;25:1054-1060.
  5. Yang CC, Huang YT, Yu CH, et al. Inflammatory facial acne during uncomplicated pregnancy and post‐partum in adult women: a preliminary hospital‐based prospective observational study of 35 cases from Taiwan. J Eur Acad Dermatol Venereol. 2016;30:1787-1789.
  6. Dréno B, Blouin E, Moyse D, et al. Acne in pregnant women: a French survey. Acta Derm Venereol. 2014;94:82-83.
  7. Kutlu Ö, Karadag˘ AS, Ünal E, et al. Acne in pregnancy: a prospective multicenter, cross‐sectional study of 295 patients in Turkey. Int J Dermatol. 2020;59:1098-1105.
  8. Hoefel IDR, Weber MB, Manzoni APD, et al. Striae gravidarum, acne, facial spots, and hair disorders: risk factors in a study with 1284 puerperal patients. J Pregnancy. 2020;2020:8036109.
  9. Ayanlowo OO, Otrofanowei E, Shorunmu TO, et al. Pregnancy dermatoses: a study of patients attending the antenatal clinic at two tertiary care centers in south west Nigeria. PAMJ Clin Med. 2020;3.
  10. Bechstein S, Ochsendorf F. Acne and rosacea in pregnancy. Hautarzt. 2017;68:111-119.
  11. Habeshian KA, Cohen BA. Current issues in the treatment of acne vulgaris. Pediatrics. 2020;145(suppl 2):S225-S230.
  12. Content and format of labeling for human prescription drug and biological products; requirements for pregnancy and lactation labeling (21 CFR 201). Fed Regist. 2014;79:72064-72103.
  13. Sagransky M, Yentzer BA, Feldman SR. Benzoyl peroxide: a review of its current use in the treatment of acne vulgaris. Expert Opin Pharmacother. 2009;10:2555-2562.
  14. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation: part I. Pregnancy. J Am Acad Dermatol. 2014;70:401.e1-401.e14; quiz 415.
  15. Wolverton SE. Systemic corticosteroids. Comprehensive Dermatol Drug Ther. 2012;3:143-168.
  16. Kirtschig G, Schaefer C. Dermatological medications and local therapeutics. In: Schaefer C, Peters P, Miller RK, eds. Drugs During Pregnancy and Lactation. 3rd edition. Elsevier; 2015:467-492.
  17. Pugashetti R, Shinkai K. Treatment of acne vulgaris in pregnant patients. Dermatol Ther. 2013;26:302-311.
  18. Touitou E, Godin B, Shumilov M, et al. Efficacy and tolerability of clindamycin phosphate and salicylic acid gel in the treatment of mild to moderate acne vulgaris. J Eur Acad Dermatol Venereol. 2008;22:629-631.
  19. Schaefer C, Peters PW, Miller RK, eds. Drugs During Pregnancy and Lactation: Treatment Options and Risk Assessment. 2nd ed. Academic Press; 2014.
  20. Birmingham B, Greene D, Rhodes C. Systemic absorption of topical salicylic acid. Int J Dermatol. 1979;18:228-231.
  21. Trivedi NA. A meta-analysis of low-dose aspirin for prevention of preeclampsia. J Postgrad Med. 2011;57:91-95.
  22. Lucky AW, Maloney JM, Roberts J, et al. Dapsone gel 5% for the treatment of acne vulgaris: safety and efficacy of long-term (1 year) treatment. J Drugs Dermatol. 2007;6:981-987.
  23. Nosten F, McGready R, d’Alessandro U, et al. Antimalarial drugs in pregnancy: a review. Curr Drug Saf. 2006;1:1-15.
  24. Brabin BJ, Eggelte TA, Parise M, et al. Dapsone therapy for malaria during pregnancy: maternal and fetal outcomes. Drug Saf. 2004;27:633-648.
  25. Tuffanelli DL. Successful pregnancy in a patient with dermatitis herpetiformis treated with low-dose dapsone. Arch Dermatol. 1982;118:876.
  26. Meredith FM, Ormerod AD. The management of acne vulgaris in pregnancy. Am J Clin Dermatol. 2013;14:351-358.
  27. Kong Y, Tey H. Treatment of acne vulgaris during pregnancy and lactation. Drugs. 2013;73:779-787.
  28. Leachman SA, Reed BR. The use of dermatologic drugs in pregnancy and lactation. Dermatol Clin. 2006;24:167-197.
  29. Ly S, Kamal K, Manjaly P, et al. Treatment of acne vulgaris during pregnancy and lactation: a narrative review. Dermatol Ther. 2023;13:115-130.
  30. Webster G. Combination azelaic acid therapy for acne vulgaris. J Am Acad Dermatol. 2000;43:S47-S50.
  31. Archer CB, Cohen SN, Baron SE. Guidance on the diagnosis and clinical management of acne. Clin Exp Dermatol. 2012;37(suppl 1):1-6.
  32. Graupe K, Cunliffe W, Gollnick H, et al. Efficacy and safety of topical azelaic acid (20 percent cream): an overview of results from European clinical trials and experimental reports. Cutis. 1996;57(1 suppl):20-35.
  33. Bozzo P, Chua-Gocheco A, Einarson A. Safety of skin care products during pregnancy. Can Fam Physician. 2011;57:665-667.
  34. Munley SM, Kennedy GL, Hurtt ME. Developmental toxicity study of glycolic acid in rats. Drug Chem Toxicol. 1999;22:569-582.
  35. Chien AL, Qi J, Rainer B, et al. Treatment of acne in pregnancy. J Am Board Fam Med. 2016;29:254-262.
  36. Stuart B, Maund E, Wilcox C, et al. Topical preparations for the treatment of mild‐to‐moderate acne vulgaris: systematic review and network meta‐analysis. Br J Dermatol. 2021;185:512-525.
  37. van Hoogdalem EJ, Baven TL, Spiegel‐Melsen I, et al. Transdermal absorption of clindamycin and tretinoin from topically applied anti‐acne formulations in man. Biopharm Drug Dispos. 1998;19:563-569.
  38. Austin BA, Fleischer AB Jr. The extinction of topical erythromycin therapy for acne vulgaris and concern for the future of topical clindamycin. J Dermatolog Treat. 2017;28:145-148.
  39. Eady EA, Cove J, Holland K, et al. Erythromycin resistant propionibacteria in antibiotic treated acne patients: association with therapeutic failure. Br J. Dermatol. 1989;121:51-57.
  40. Alkhawaja E, Hammadi S, Abdelmalek M, et al. Antibiotic resistant Cutibacterium acnes among acne patients in Jordan: a cross sectional study. BMC Dermatol. 2020;20:1-9.
  41. Han G, Wu JJ, Del Rosso JQ. Use of topical tazarotene for the treatment of acne vulgaris in pregnancy: a literature review. J Clin Aesthet Dermatol. 2020;13:E59-E65.
  42. Selcen D, Seidman S, Nigro MA. Otocerebral anomalies associated with topical tretinoin use. Brain Dev. 2000;22:218-220.
  43. Moretz D. Drug Class Update with New Drug Evaluations: Topical Products for Inflammatory Skin Conditions. Oregon State University Drug Use & Research Management Program; December 2022. Accessed January 8, 2024. https://www.orpdl.org/durm/meetings/meetingdocs/2022_12_01/archives/2022_12_01_Inflammatory_Skin_Dz_ClassUpdate.pdf
  44. Kaplan YC, Ozsarfati J, Etwel F, et al. Pregnancy outcomes following first‐trimester exposure to topical retinoids: a systematic review and meta‐analysis. Br J Dermatol. 2015;173:1132-1141.
  45. Menter A. Pharmacokinetics and safety of tazarotene. J Am Acad Dermatol. 2000;43(2, pt 3):S31-S35.
  46. Autret E, Berjot M, Jonville-Béra A-P, et al. Anophthalmia and agenesis of optic chiasma associated with adapalene gel in early pregnancy. Lancet. 1997;350:339.
  47. Weiss J, Mallavalli S, Meckfessel M, et al. Safe use of adapalene 0.1% gel in a non-prescription environment. J Drugs Dermatol. 2021;20:1330-1335.
  48. Alessandro Mazzetti M. A phase 2b, randomized, double-blind vehicle controlled, dose escalation study evaluating clascoterone 0.1%, 0.5%, and 1% topical cream in subjects with facial acne. J Drugs Dermatol. 2019;18:570-575.
  49. Eichenfield L, Hebert A, Gold LS, et al. Open-label, long-term extension study to evaluate the safety of clascoterone (CB-03-01) cream, 1% twice daily, in patients with acne vulgaris. J Am Acad Dermatol. 2020;83:477-485.
  50. Trifu V, Tiplica GS, Naumescu E, et al. Cortexolone 17α‐propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. a pilot randomized, double‐blind comparative study vs. placebo and tretinoin 0.05% cream. Br J Dermatol. 2011;165:177-183.
  51. Hebert A, Thiboutot D, Gold LS, et al. Efficacy and safety of topical clascoterone cream, 1%, for treatment in patients with facial acne: two phase 3 randomized clinical trials. JAMA Dermatol. 2020;156:621-630.
  52. Alkhodaidi ST, Al Hawsawi KA, Alkhudaidi IT, et al. Efficacy and safety of topical clascoterone cream for treatment of acne vulgaris: a systematic review and meta‐analysis of randomized placebo‐controlled trials. Dermatol Ther. 2021;34:e14609.
  53. Clasoterone. Package insert. Cassiopea Inc; 2020.
  54. Paik J. Topical minocycline foam 4%: a review in acne vulgaris. Am J Clin Dermatol. 2020;21:449-456.
  55. Jones TM, Ellman H. Pharmacokinetic comparison of once-daily topical minocycline foam 4% vs oral minocycline for moderate-to-severe acne. J Drugs Dermatol. 2017;16:1022-1028.
  56. Minocycline hydrochloride extended-release tablets. Package insert. JG Pharma; July 2020. Accessed January 8, 2024. https://www.jgpharmainc.com/assets/pdf/minocycline-hydrochloride.pdf
  57. Dinnendahl V, Fricke U (eds). Arzneistoff-Profile: Basisinformation über arzneiliche Wirkstoffe. Govi Pharmazeutischer Verlag; 2010.
  58. Martins AM, Marto JM, Johnson JL, et al. A review of systemic minocycline side effects and topical minocycline as a safer alternative for treating acne and rosacea. Antibiotics. 2021;10:757.
  59. Landis MN. Optimizing isotretinoin treatment of acne: update on current recommendations for monitoring, dosing, safety, adverse effects, compliance, and outcomes. Am J Clin Dermatol. 2020;21:411-419.
  60. Draghici C-C, Miulescu R-G, Petca R-C, et al. Teratogenic effect of isotretinoin in both fertile females and males. Exp Ther Med. 2021;21:1-5.
  61. Barker RA, Wilcox C, Layton AM. Oral spironolactone for acne vulgaris in adult females: an update of the literature. Am J Clin Dermatol. 2020;21:303-305.
  62. Han JJ, Faletsky A, Barbieri JS, et al. New acne therapies and updates on use of spironolactone and isotretinoin: a narrative review. Dermatol Ther (Heidelb). 2021;11:79-91.
  63. Briggs GG, Freeman RK, Yaffe SJ. Drugs in Pregnancy and Lactation: A Reference Guide to Fetal and Neonatal Risk. Lippincott Williams & Wilkins; 2012.
  64. Patel DJ, Bhatia N. Oral antibiotics for acne. Am J Clin Dermatol. 2021;22:193-204.
  65. Jick H, Holmes LB, Hunter JR, et al. First-trimester drug use and congenital disorders. JAMA. 1981;246:343-346.
  66. Valente Duarte de Sousa IC. An overview of sarecycline for the treatment of moderate-to-severe acne vulgaris. Exp Opin Pharmacother. 2021;22:145-154.
  67. Hussar DA, Chahine EB. Omadacycline tosylate, sarecycline hydrochloride, rifamycin sodium, and moxidectin. J Am Pharm Assoc. 2019;59:756-760.
  68. Haidari W, Bruinsma R, Cardenas-de la Garza JA, et al. Sarecycline review. Ann Pharmacother. 2020;54:164-170.
  69. Feldman S, Careccia RE, Barham KL, et al. Diagnosis and treatment of acne. Am Fam Physician. 2004;69:2123-2130.
  70. Gammon WR, Meyer C, Lantis S, et al. Comparative efficacy of oral erythromycin versus oral tetracycline in the treatment of acne vulgaris: a double-blind study. J Am Acad Dermatol. 1986;14:183-186.
  71. Källén BA, Olausson PO, Danielsson BR. Is erythromycin therapy teratogenic in humans? Reprod Toxicol. 2005;20:209-214.
  72. McCormack WM, George H, Donner A, et al. Hepatotoxicity of erythromycin estolate during pregnancy. Antimicrob Agents Chemother. 1977;12:630-635.
  73. Cervantes J, Eber AE, Perper M, et al. The role of zinc in the treatment of acne: a review of the literature. Dermatolog Ther. 2018;31:e12576.
  74. Dréno B, Blouin E. Acne, pregnant women and zinc salts: a literature review [in French]. Ann Dermatol Venereol. 2008;135:27-33.
  75. Eid MM, Saleh MS, Allam NM, et al. Narrow band ultraviolet B versus red light-emitting diodes in the treatment of facial acne vulgaris: a randomized controlled trial. Photobiomodul Photomed Laser Surg. 2021;39:418-424.
  76. Zeichner JA. Narrowband UV-B phototherapy for the treatment of acne vulgaris during pregnancy. Arch Dermatol. 2011;147:537-539.
  77. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. Lasers Med Sci. 2011;26:481-485.
  78. Park KK, Murase JE. Narrowband UV-B phototherapy during pregnancy and folic acid depletion. Arch Dermatol. 2012;148:132-133.
  79. Jablonski NG. A possible link between neural tube defects and ultraviolet light exposure. Med Hypotheses. 1999;52:581-582.
  80. Zhang M, Goyert G, Lim HW. Folate and phototherapy: what should we inform our patients? J Am Acad Dermatol. 2017;77:958-964.
  81. AviClear. Cutera website. Accessed January 8, 2024. https://www.cutera.com/solutions/aviclear/
  82. Wu X, Yang Y, Wang Y, et al. Treatment of refractory acne using selective sebaceous gland electro-thermolysis combined with non-thermal plasma. J Cosmet Laser Ther. 2021;23:188-194.
  83. Ahn GR, Kim JM, Park SJ, et al. Selective sebaceous gland electrothermolysis using a single microneedle radiofrequency device for acne patients: a prospective randomized controlled study. Lasers Surg Med. 2020;52:396-401.
  84. Fabbrocini G, De Padova MP, Tosti A. Chemical peels: what’s new and what isn’t new but still works well. Facial Plast Surg. 2009;25:329-336.
  85. Andersen FA. Final report on the safety assessment of glycolic acid, ammonium, calcium, potassium, and sodium glycolates, methyl, ethyl, propyl, and butyl glycolates, and lactic acid, ammonium, calcium, potassium, sodium, and TEA-lactates, methyl, ethyl, isopropyl, and butyl lactates, and lauryl, myristyl, and cetyl lactates. Int J Toxicol. 1998;17(1_suppl):1-241.
  86. Lee KC, Korgavkar K, Dufresne RG Jr, et al. Safety of cosmetic dermatologic procedures during pregnancy. Dermatol Surg. 2013;39:1573-1586.
  87. James AH, Brancazio LR, Price T. Aspirin and reproductive outcomes. Obstet Gynecol Surv. 2008;63:49-57.
  88. Zhou W-S, Xu L, Xie S-H, et al. Decreased birth weight in relation to maternal urinary trichloroacetic acid levels. Sci Total Environ. 2012;416:105-110.
  89. Schwartz DB, Greenberg MD, Daoud Y, et al. Genital condylomas in pregnancy: use of trichloroacetic acid and laser therapy. Am J Obstet Gynecol. 1988;158:1407-1416.
  90. Starkman SJ, Mangat DS. Chemical peel (deep, medium, light). Facial Plast Surg Clin North Am. 2020;28:45-57.
  91. Trivedi M, Kroumpouzos G, Murase J. A review of the safety of cosmetic procedures during pregnancy and lactation. Int J Womens Dermatol. 2017;3:6-10.
  92. Gallagher T, Taliercio M, Nia JK, et al. Dermatologist use of intralesional triamcinolone in the treatment of acne. J Clin Aesthet Dermatol. 2020;13:41-43.
  93. Zamil DH, Burns EK, Perez-Sanchez A, et al. Risk of birth defects from vitamin A “acne supplements” sold online. Dermatol Pract Concept. 2021;11:e2021075.
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
E26-E32
Page Number
E26-E32
Publications
Publications
Topics
Article Type
Display Headline
Acne and Pregnancy: A Clinical Review and Practice Pearls
Display Headline
Acne and Pregnancy: A Clinical Review and Practice Pearls
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Yaghi</fileName> <TBEID>0C02F172.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F172</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Acne and Pregnancy</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240125T100045</firstPublished> <LastPublished>20240125T100045</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240125T100045</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Yaghi</byline> <bylineText>Marita Yaghi, MD; Daniela Baboun, BA; Jonette E. Keri, MD, PhD </bylineText> <bylineFull>Yaghi</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E26-E32</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Acne vulgaris, or acne, is a highly common inflammatory skin disorder affecting up to 85% of the population, and it constitutes the most commonly presenting chi</metaDescription> <articlePDF>300038</articlePDF> <teaserImage/> <title>Acne and Pregnancy: A Clinical Review and Practice Pearls</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2293</CMSID> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>acne</keyword> <keyword> pregnancy</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Original Article | 2293<pubSubsection/></pubSection> </pubSections> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">171</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026ac.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Acne and Pregnancy: A Clinical Review and Practice Pearls</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Acne vulgaris is a common condition that routinely affects females of childbearing age. Taking into consideration the reproductive journey of women when treating acne is of paramount importance given the safety concerns to both the mother and the fetus associated with certain medications. Therefore, careful consideration of therapeutic choices during pregnancy is crucial. Herein, we summarize the safety of acne treatments during pregnancy and offer practical clinical pearls for routine dermatology practice. </p> <p> <em><em>Cutis.</em> 2024;113:E26-E32.</em> </p> <p>Acne vulgaris, or acne, is a highly common inflammatory skin disorder affecting up to 85% of the population, and it constitutes the most commonly presenting chief concern in routine dermatology practice.<sup>1</sup> Older teenagers and young adults are most often affected by acne.<sup>2</sup> Although acne generally is more common in males, adult-onset acne occurs more frequently in women.<sup>2,3</sup> Black and Hispanic women are at higher risk for acne compared to those of Asian, White, or Continental Indian descent.<sup>4</sup> As such, acne is a common concern in all women of childbearing age. </p> <p>Concerns for maternal and fetal safety are important therapeutic considerations, especially because hormonal and physiologic changes in pregnancy can lead to onset of inflammatory acne lesions, particularly during the second and third trimesters.<sup>5</sup> Female patients younger than 25 years; with a higher body mass index, prior irregular menstruation, or polycystic ovary syndrome; or those experiencing their first pregnancy are thought to be more commonly affected.<sup>5-7</sup> In fact, acne affects up to 43% of pregnant women, and lesions typically extend beyond the face to involve the trunk.<sup>6,8-10</sup> Importantly, one-third of women with a history of acne experience symptom relapse after disease-free periods, while two-thirds of those with ongoing disease experience symptom deterioration during pregnancy.<sup>10</sup> Although acne is not a life-threatening condition, it has a well-documented, detrimental impact on social, emotional, and psychological well-being, namely self-perception, social interactions, quality-of-life scores, depression, and anxiety.<sup>11<br/><br/></sup>Therefore, safe and effective treatment of pregnant women is of paramount importance. Because pregnant women are not included in clinical trials, there is a paucity of medication safety data, further augmented by inefficient access to available information. The US Food and Drug Administration (FDA) pregnancy safety categories were updated in 2015, letting go of the traditional A, B, C, D, and X categories.<sup>12</sup> The Table reviews the current pregnancy classification system. In this narrative review, we summarize the most recent available data and recommendations on the safety and efficacy of acne treatment during pregnancy. </p> <h3>Topical Treatments for Acne</h3> <p><span class="sub3">Benzoyl Peroxide—</span>Benzoyl peroxide commonly is used as first-line therapy alone or in combination with other agents for the treatment of mild to moderate acne.<sup>13</sup> It is safe for use during pregnancy.<sup>14</sup> Although the medication is systemically absorbed, it undergoes complete metabolism to benzoic acid, a commonly used food additive.<sup>15,16</sup> Benzoic acid has low bioavailability, as it gets rapidly metabolized by the kidneys; therefore, benzoyl peroxide is unlikely to reach clinically significant levels in the maternal circulation and consequently the fetal circulation. Additionally, it has a low risk for causing congenital malformations.<sup>17</sup></p> <p><span class="sub3">Salicylic Acid—</span>For mild to moderate acne, salicylic acid is a second-line agent that likely is safe for use by pregnant women at low concentrations and over limited body surface areas.<sup>14,18,19</sup> There is minimal systemic absorption of the drug.<sup>20</sup> Additionally, aspirin, which is broken down in the body into salicylic acid, is used in low doses for the treatment of pre-eclampsia during pregnancy.<sup>21<br/><br/></sup><span class="sub3">Dapsone—</span>The use of dapsone gel 5% as a second-line agent has shown efficacy for mild to moderate acne.<sup>22</sup> The oral formulation, commonly used for malaria and leprosy prophylaxis, has failed to show associated fetal toxicity or congenital anomalies.<sup>23,24</sup> It also has been used as a first-line treatment for dermatitis herpetiformis in pregnancy.<sup>25</sup> Although the medication likely is safe, it is better to minimize its use during the third trimester to reduce the theoretical risk for hyperbilirubinemia in the neonate.<sup>17,26-29<br/><br/></sup><span class="sub3">Azelaic Acid—</span>Azelaic acid effectively targets noninflammatory and inflammatory acne and generally is well tolerated, harboring a good safety profile.<sup>30</sup> Topical 20% azelaic acid has localized antibacterial and comedolytic effects and is safe for use during pregnancy.<sup>31,32<br/><br/></sup><span class="sub3">Glycolic Acid—</span>Limited data exist on the safety of glycolic acid during pregnancy. In vitro studies have shown up to 27% systemic absorption depending on pH, concentration, and duration of application.<sup>33</sup> Animal reproductive studies involving rats have shown fetal multisystem malformations and developmental abnormalities with oral administration of glycolic acid at doses far exceeding those used in humans.<sup>34</sup> Although no human reproductive studies exist, topical glycolic acid is unlikely to reach the developing fetus in notable amounts, and the medication is likely safe for use.<sup>17,35<br/><br/></sup><span class="sub3">Clindamycin—</span>Topical clindamycin phosphate is an effective and well-tolerated agent for the treatment of mild to moderate acne.<sup>36</sup> Its systemic absorption is minimal, and it is considered safe for use during all trimesters of pregnancy.<sup>14,17,26,27,35,37<br/><br/></sup><span class="sub3">Erythromycin—</span>Topical erythromycin is another commonly prescribed topical antibiotic used to target mild to moderate acne. However, its use recently has been associated with a decrease in efficacy secondary to the rise of antibacterial resistance in the community.<sup>38-40</sup> Nevertheless, it remains a safe treatment for use during all trimesters of pregnancy.<sup>14,17,26,27,35,37<br/><br/></sup><span class="sub3">Topical Retinoids—</span>Vitamin A derivatives (also known as retinoids) are the mainstay for the treatment of mild to moderate acne. Limited data exist regarding pregnancy outcomes after in utero exposure.<sup>41</sup> A rare case report suggested topical tretinoin has been associated with fetal otocerebral anomalies.<sup>42</sup> For tazarotene, teratogenic effects were seen in animal reproductive studies at doses exceeding maximum recommended human doses.<sup>41,43</sup> However, a large meta-analysis failed to find a clear risk for increased congenital malformations, spontaneous abortions, stillbirth, elective termination of pregnancy, low birthweight, or prematurity following first-trimester exposure to topical retinoids.<sup>44</sup> As the level of exposure that could lead to teratogenicity in humans is unknown, avoidance of both tretinoin and tazarotene is recommended in pregnant women.<sup>41,45</sup> Nevertheless, women inadvertently exposed should be reassured.<sup>44<br/><br/></sup>Conversely, adapalene has been associated with 1 case of anophthalmia and agenesis of the optic chiasma in a fetus following exposure until 13 weeks’ gestation.<sup>46</sup> However, a large, open-label trial prior to the patient transitioning from adapalene to over-the-counter treatment showed that the drug harbors a large and reassuring margin of safety and no risk for teratogenicity in a maximal usage trial and Pregnancy Safety Review.<sup>47</sup> Therefore, adapalene gel 0.1% is a safe and effective medication for the treatment of acne in a nonprescription environment and does not pose harm to the fetus.<br/><br/><span class="sub3">Clascoterone—</span>Clascoterone is a novel topical antiandrogenic drug approved for the treatment of hormonal and inflammatory moderate to severe acne.<sup>48-51</sup> Human reproductive data are limited to 1 case of pregnancy that occurred during phase 3 trial investigations, and no adverse outcomes were reported.<sup>51</sup> Minimal systemic absorption follows topical use.<sup>52</sup> Nonetheless, dose-independent malformations were reported in animal reproductive studies.<sup>53</sup> As such, it remains better to avoid the use of clascoterone during pregnancy pending further safety data.<br/><br/><span class="sub3">Minocycline Foam—</span>Minocycline foam 4% is approved to treat inflammatory lesions of nonnodular moderate to severe acne in patients 9 years and older.<sup>54</sup> Systemic absorption is minimal, and the drug has limited bioavailability with minimal systemic accumulation in the patient’s serum.<sup>55</sup> Given this information, it is unlikely that topical minocycline will reach notable levels in the fetal serum or harbor teratogenic effects, as seen with the oral formulation.<sup>56</sup> However, it may be best to avoid its use during the second and third trimesters given the potential risk for tooth discoloration in the fetus.<sup>57,58</sup></p> <h3>Systemic Treatments for Acne</h3> <p><span class="sub3">Isotretinoin—</span>Isotretinoin is the most effective treatment for moderate to severe acne with a well-documented potential for long-term clearance.<sup>59</sup> Its use during pregnancy is absolutely contraindicated, as the medication is a well-known teratogen. Associated congenital malformations include numerous craniofacial defects, cardiovascular and neurologic malformations, or thymic disorders that are estimated to affect 20% to 35% of infants exposed in utero.<sup>60</sup> Furthermore, strict contraception use during treatment is mandated for patients who can become pregnant. It is recommended to wait at least 1 month and 1 menstrual cycle after medication discontinuation before attempting to conceive.<sup>17</sup> Pregnancy termination is recommended if conception occurs during treatment with isotretinoin. </p> <p><span class="sub3">Spironolactone—</span>Spironolactone is an androgen-receptor antagonist commonly prescribed off label for mild to severe acne in females.<sup>61,62</sup> Spironolactone promotes the feminization of male fetuses and should be avoided in pregnancy.<sup>63</sup> <br/><br/><span class="sub3">Doxycycline/Minocycline—</span>Tetracyclines are the most commonly prescribed oral antibiotics for moderate to severe acne.<sup>64</sup> Although highly effective at treating acne, tetracyclines generally should be avoided in pregnancy. First-trimester use of doxycycline is not absolutely contraindicated but should be reserved for severe illness and not employed for the treatment of acne. However, accidental exposure to doxycycline has not been associated with congenital malformations.<sup>65</sup> Nevertheless, after the 15th week of gestation, permanent tooth discoloration and bone growth inhibition in the fetus are serious and well-documented risks.<sup>14,17</sup> Additional adverse events following in utero exposure include infantile inguinal hernia, hypospadias, and limb hypoplasia.<sup>63<br/><br/></sup><span class="sub3">Sarecycline—</span>Sarecycline is a novel tetracycline-class antibiotic for the treatment of moderate to severe inflammatory acne. It has a narrower spectrum of activity compared to its counterparts within its class, which translates to an improved safety profile, namely when it comes to gastrointestinal tract microbiome disruption and potentially decreased likelihood of developing bacterial resistance.<sup>66</sup> Data on human reproductive studies are limited, but it is advisable to avoid sarecycline in pregnancy, as it may cause adverse developmental effects in the fetus, such as reduced bone growth, in addition to the well-known tetracycline-associated risk for permanent discoloration of the teeth if used during the second and third trimesters.<sup>67,68<br/><br/></sup><span class="sub3">Erythromycin—</span>Oral erythromycin targets moderate to severe inflammatory acne and is considered safe for use during pregnancy.<sup>69,70</sup> There has been 1 study reporting an increased risk for atrial and ventricular septal defects (1.8%) and pyloric stenosis (0.2%), but these risks are still uncertain, and erythromycin is considered compatible with pregnancy.<sup>71</sup> However, erythromycin estolate formulations should be avoided given the associated 10% to 15% risk for reversible cholestatic liver injury.<sup>72</sup> Erythromycin base or erythromycin ethylsuccinate formulations should be favored. <br/><br/><span class="sub3">Systemic Steroids—</span>Prednisone is indicated for severe acne with scarring and should only be used during pregnancy after clearance from the patient’s obstetrician. Doses of 0.5 mg/kg or less should be prescribed in combination with systemic antibiotics as well as agents for bone and gastrointestinal tract prophylaxis.<sup>29<br/><br/></sup><span class="sub3">Zinc—</span>The exact mechanism by which zinc exerts its effects to improve acne remains largely obscure. It has been found effective against inflammatory lesions of mild to moderate acne.<sup>73</sup> Generally recommended dosages range from 30 to 200 mg/d but may be associated with gastrointestinal tract disturbances. Dosages of 75 mg/d have shown no harm to the fetus.<sup>74</sup> When taking this supplement, patients should not exceed the recommended doses given the risk for hypocupremia associated with high-dose zinc supplementation. </p> <h3>Light-Based Therapies </h3> <p><span class="sub3">Phototherapy—</span>Narrowband UVB phototherapy is effective for the treatment of mild to moderate acne.<sup>75</sup> It has been proven to be a safe treatment option during pregnancy, but its use has been associated with decreased folic acid levels.<sup>76-79</sup> Therefore, in addition to attaining baseline folic acid serum levels, supplementation with folic acid prior to treatment, as per routine prenatal guidelines, should be sought.<sup>80</sup></p> <p><span class="sub3">AviClear—</span>The AviClear (Cutera) laser is the first device cleared by the FDA for mild to severe acne in March 2022.<sup>81</sup> The FDA clearance for the Accure (Accure Acne Inc) laser, also targeting mild to severe acne, followed soon after (November 2022). Both lasers harbor a wavelength of 1726 nm and target sebaceous glands with electrothermolysis.<sup>82,83</sup> Further research and long-term safety data are required before using them in pregnancy.</p> <h3>Other Therapies </h3> <p><span class="sub3">Cosmetic Peels—</span>Glycolic acid peels induce epidermolysis and desquamation.<sup>84</sup> Although data on use during pregnancy are limited, these peels have limited dermal penetration and are considered safe for use in pregnancy.<sup>33,85,86</sup> Similarly, keratolytic lactic acid peels harbor limited dermal penetration and can be safely used in pregnant women.<sup>87-89</sup> Salicylic acid peels also work through epidermolysis and desquamation<sup>84</sup>; however, they tend to penetrate deeper into the skin, reaching down to the basal layer, if large areas are treated or when applied under occlusion.<sup>86,90</sup> Although their use is not contraindicated in pregnancy, they should be limited to small areas of coverage.<sup>91</sup> </p> <p><span class="sub3">Intralesional Triamcinolone—</span>Acne cysts and inflammatory papules can be treated with intralesional triamcinolone injections to relieve acute symptoms such as pain.<sup>92</sup> Low doses at concentrations of 2.5 mg/mL are considered compatible with pregnancy when indicated.<sup>29</sup></p> <h3>Approaching the Patient Clinical Encounter </h3> <p>In patients seeking treatment prior to conception, a few recommendations can be made to minimize the risk for acne recurrence or flares during pregnancy. For instance, because data show an association between increased acne severity in those with a higher body mass index and in pregnancy, weight loss may be recommended prior to pregnancy to help mitigate symptoms after conception.<sup>7</sup> The Figure summarizes our recommendations for approaching and treating acne in pregnancy.</p> <p>In all patients, grading the severity of the patient’s acne as mild, moderate, or severe is the first step. The presence of scarring is an additional consideration during the physical examination and should be documented. A careful discussion of treatment expectations and prognosis should be the focus before treatment initiation. Meticulous documentation of the physical examination and discussion with the patient should be prioritized.<br/><br/>To minimize toxicity and risks to the developing fetus, monotherapy is favored. Topical therapy should be considered first line. Safe regimens include mild nonabrasive washes, such as those containing benzoyl peroxide or glycolic acid, or topical azelaic acid or clindamycin phosphate for mild to moderate acne. More severe cases warrant the consideration of systemic medications as second line, as more severe acne is better treated with oral antibiotics such as the macrolides erythromycin or clindamycin or systemic corticosteroids when concern exists for severe scarring. The additional use of physical sunscreen also is recommended. <br/><br/>An important topic to address during the clinical encounter is cautious intake of oral supplements for acne during pregnancy, as they may contain harmful and teratogenic ingredients. A recent search focusing on acne supplements available online between March and May 2020 uncovered 49 different supplements, 26 (53%) of which contained vitamin A.<sup>93</sup> Importantly, 3 (6%) of these 49 supplements were likely teratogenic, 4 (8%) contained vitamin A doses exceeding the recommended daily nutritional intake level, and 15 (31%) harbored an unknown teratogenic risk. Furthermore, among the 6 (12%) supplements with vitamin A levels exceeding 10,000 IU, 2 lacked any mention of pregnancy warning, including the supplement with the highest vitamin A dose found in this study.<sup>93</sup> Because dietary supplements are not subject to the same stringent regulations by the FDA as drugs, inadvertent use by unaware patients ought to be prevented by careful counseling and education.<br/><br/>Finally, patients should be counseled to seek care following delivery for potentially updated medication management of acne, especially if they are breastfeeding. Co-management with a pediatrician may be indicated during lactation, particularly when newborns are born preterm or with other health conditions that may warrant additional caution with the use of certain agents.</p> <h2>References </h2> <p class="reference"> 1. Bhate K, Williams H. Epidemiology of acne vulgaris. <i>Br J Dermatol</i>. 2013;168:474-485. <br/><br/> 2. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. <i>Sci Rep</i>. 2020;10:5754. <br/><br/> 3. Fisk WA, Lev-Tov HA, Sivamani RK. Epidemiology and management of acne in adult women. <i>Curr Dermatol Rep</i>. 2014;3:29-39. <br/><br/> 4. Perkins A, Cheng C, Hillebrand G, et al. Comparison of the epidemiology of acne vulgaris among Caucasian, Asian, Continental Indian and African American women. <i>J Eur Acad Dermatol Venereol</i>. 2011;25:1054-1060. <br/><br/> 5. Yang CC, Huang YT, Yu CH, et al. Inflammatory facial acne during uncomplicated pregnancy and post‐partum in adult women: a preliminary hospital‐based prospective observational study of 35 cases from Taiwan. <i>J Eur Acad Dermatol Venereol</i>. 2016;30:1787-1789. <br/><br/> 6. Dréno B, Blouin E, Moyse D, et al. Acne in pregnant women: a French survey. <i>Acta Derm Venereol</i>. 2014;94:82-83. <br/><br/> 7. Kutlu Ö, Karadag˘ AS, Ünal E, et al. Acne in pregnancy: a prospective multicenter, cross‐sectional study of 295 patients in Turkey. <i>Int J Dermatol</i>. 2020;59:1098-1105. <br/><br/> 8. Hoefel IDR, Weber MB, Manzoni APD, et al. Striae gravidarum, acne, facial spots, and hair disorders: risk factors in a study with 1284 puerperal patients. <i>J Pregnancy</i>. 2020;2020:8036109. <br/><br/> 9. Ayanlowo OO, Otrofanowei E, Shorunmu TO, et al. Pregnancy dermatoses: a study of patients attending the antenatal clinic at two tertiary care centers in south west Nigeria. <i>PAMJ Clin Med</i>. 2020;3.<br/><br/>10. Bechstein S, Ochsendorf F. Acne and rosacea in pregnancy. <i>Hautarzt</i>. 2017;68:111-119. <br/><br/>11. Habeshian KA, Cohen BA. Current issues in the treatment of acne vulgaris. <i>Pediatrics</i>. 2020;145(suppl 2):S225-S230. <br/><br/>12. Content and format of labeling for human prescription drug and biological products; requirements for pregnancy and lactation labeling (21 CFR 201). <i>Fed Regist</i>. 2014;79:72064-72103. <br/><br/>13. Sagransky M, Yentzer BA, Feldman SR. Benzoyl peroxide: a review of its current use in the treatment of acne vulgaris. <i>Expert Opin Pharmacother</i>. 2009;10:2555-2562. <br/><br/>14. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation: part I. Pregnancy. <i>J Am Acad Dermatol</i>. 2014;70:401.e1-401.e14; quiz 415. <br/><br/>15. Wolverton SE. Systemic corticosteroids. <i>Comprehensive Dermatol Drug Ther</i>. 2012;3:143-168. <br/><br/>16. Kirtschig G, Schaefer C. Dermatological medications and local therapeutics. In: Schaefer C, Peters P, Miller RK, eds. <i>Drugs During Pregnancy and Lactation</i>. 3rd edition. Elsevier; 2015:467-492.<br/><br/>17. Pugashetti R, Shinkai K. Treatment of acne vulgaris in pregnant patients. <i>Dermatol Ther</i>. 2013;26:302-311. <br/><br/>18. Touitou E, Godin B, Shumilov M, et al. Efficacy and tolerability of clindamycin phosphate and salicylic acid gel in the treatment of mild to moderate acne vulgaris. <i>J Eur Acad Dermatol Venereol</i>. 2008;22:629-631. <br/><br/>19. Schaefer C, Peters PW, Miller RK, eds. <i>Drugs During Pregnancy and Lactation: Treatment Options and Risk Assessment</i>. 2nd ed. Academic Press; 2014. <br/><br/>20. Birmingham B, Greene D, Rhodes C. Systemic absorption of topical salicylic acid. <i>Int J Dermatol</i>. 1979;18:228-231. <br/><br/>21. Trivedi NA. A meta-analysis of low-dose aspirin for prevention of preeclampsia. <i>J Postgrad Med</i>. 2011;57:91-95. <br/><br/>22. Lucky AW, Maloney JM, Roberts J, et al. Dapsone gel 5% for the treatment of acne vulgaris: safety and efficacy of long-term (1 year) treatment. <i>J Drugs Dermatol</i>. 2007;6:981-987. <br/><br/>23. Nosten F, McGready R, d’Alessandro U, et al. Antimalarial drugs in pregnancy: a review. <i>Curr Drug Saf</i>. 2006;1:1-15. <br/><br/>24. Brabin BJ, Eggelte TA, Parise M, et al. Dapsone therapy for malaria during pregnancy: maternal and fetal outcomes. <i>Drug Saf</i>. 2004;27:633-648. <br/><br/>25. Tuffanelli DL. Successful pregnancy in a patient with dermatitis herpetiformis treated with low-dose dapsone. <i>Arch Dermatol</i>. 1982;118:876. <br/><br/>26. Meredith FM, Ormerod AD. The management of acne vulgaris in pregnancy. <i>Am J Clin Dermatol</i>. 2013;14:351-358. <br/><br/>27. Kong Y, Tey H. Treatment of acne vulgaris during pregnancy and lactation. <i>Drugs</i>. 2013;73:779-787. <br/><br/>28. Leachman SA, Reed BR. The use of dermatologic drugs in pregnancy and lactation. <i>Dermatol Clin</i>. 2006;24:167-197. <br/><br/>29. Ly S, Kamal K, Manjaly P, et al. Treatment of acne vulgaris during pregnancy and lactation: a narrative review. <i>Dermatol Ther</i>. 2023;13:115-130. <br/><br/>30. Webster G. Combination azelaic acid therapy for acne vulgaris. <i>J Am Acad Dermatol</i>. 2000;43:S47-S50. <br/><br/>31. Archer CB, Cohen SN, Baron SE. Guidance on the diagnosis and clinical management of acne. <i>Clin Exp Dermatol</i>. 2012;37(suppl 1):1-6. <br/><br/>32. Graupe K, Cunliffe W, Gollnick H, et al. Efficacy and safety of topical azelaic acid (20 percent cream): an overview of results from European clinical trials and experimental reports. <i>Cutis</i>. 1996;57(1 suppl):20-35. </p> <p class="reference">33. Bozzo P, Chua-Gocheco A, Einarson A. Safety of skin care products during pregnancy. <i>Can Fam Physician</i>. 2011;57:665-667. <br/><br/>34. Munley SM, Kennedy GL, Hurtt ME. Developmental toxicity study of glycolic acid in rats. <i>Drug Chem Toxicol</i>. 1999;22:569-582. <br/><br/>35. Chien AL, Qi J, Rainer B, et al. Treatment of acne in pregnancy. <i>J Am Board Fam Med</i>. 2016;29:254-262. <br/><br/>36. Stuart B, Maund E, Wilcox C, et al. Topical preparations for the treatment of mild‐to‐moderate acne vulgaris: systematic review and network meta‐analysis. <i>Br J Dermatol</i>. 2021;185:512-525. 
37. van Hoogdalem EJ, Baven TL, Spiegel‐Melsen I, et al. Transdermal absorption of clindamycin and tretinoin from topically applied anti‐acne formulations in man. <i>Biopharm Drug Dispos</i>. 1998;19:563-569. <br/><br/>38. Austin BA, Fleischer AB Jr. The extinction of topical erythromycin therapy for acne vulgaris and concern for the future of topical clindamycin. <i>J Dermatolog Treat</i>. 2017;28:145-148. <br/><br/>39. Eady EA, Cove J, Holland K, et al. Erythromycin resistant propionibacteria in antibiotic treated acne patients: association with therapeutic failure. <i>Br J. Dermatol</i>. 1989;121:51-57. <br/><br/>40. Alkhawaja E, Hammadi S, Abdelmalek M, et al. Antibiotic resistant Cutibacterium acnes among acne patients in Jordan: a cross sectional study. <i>BMC Dermatol</i>. 2020;20:1-9. <br/><br/>41. Han G, Wu JJ, Del Rosso JQ. Use of topical tazarotene for the treatment of acne vulgaris in pregnancy: a literature review. <i>J Clin Aesthet Dermatol</i>. 2020;13:E59-E65. <br/><br/>42. Selcen D, Seidman S, Nigro MA. Otocerebral anomalies associated with topical tretinoin use. <i>Brain Dev</i>. 2000;22:218-220. <br/><br/>43. Moretz D. <i>Drug</i> <i>Class Update with New Drug Evaluations: Topical Products for Inflammatory Skin Conditions</i>. Oregon State University Drug Use &amp; Research Management Program; December 2022. Accessed January 8, 2024. https://www.orpdl.org/durm/meetings/meetingdocs/2022_12_01/archives/2022_12_01_Inflammatory_Skin_Dz_ClassUpdate.pdf <br/><br/>44. Kaplan YC, Ozsarfati J, Etwel F, et al. Pregnancy outcomes following first‐trimester exposure to topical retinoids: a systematic review and meta‐analysis. <i>Br J Dermatol</i>. 2015;173:1132-1141. <br/><br/>45. Menter A. Pharmacokinetics and safety of tazarotene. <i>J Am Acad Dermatol</i>. 2000;43(2, pt 3):S31-S35. <br/><br/>46. Autret E, Berjot M, Jonville-Béra A-P, et al. Anophthalmia and agenesis of optic chiasma associated with adapalene gel in early pregnancy. <i>Lancet</i>. 1997;350:339. <br/><br/>47. Weiss J, Mallavalli S, Meckfessel M, et al. Safe use of adapalene 0.1% gel in a non-prescription environment. <i>J Drugs Dermatol</i>. 2021;20:1330-1335. <br/><br/>48. Alessandro Mazzetti M. A phase 2b, randomized, double-blind vehicle controlled, dose escalation study evaluating clascoterone 0.1%, 0.5%, and 1% topical cream in subjects with facial acne. <i>J Drugs Dermatol</i>. 2019;18:570-575. <br/><br/>49. Eichenfield L, Hebert A, Gold LS, et al. Open-label, long-term extension study to evaluate the safety of clascoterone (CB-03-01) cream, 1% twice daily, in patients with acne vulgaris. <i>J Am Acad Dermatol</i>. 2020;83:477-485. <br/><br/>50. Trifu V, Tiplica GS, Naumescu E, et al. Cortexolone 17<span class="body">α</span>‐propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. a pilot randomized, double‐blind comparative study vs. placebo and tretinoin 0.05% cream. <i>Br J Dermatol</i>. 2011;165:177-183. <br/><br/>51. Hebert A, Thiboutot D, Gold LS, et al. Efficacy and safety of topical clascoterone cream, 1%, for treatment in patients with facial acne: two phase 3 randomized clinical trials. <i>JAMA Dermatol</i>. 2020;156:621-630. <br/><br/>52. Alkhodaidi ST, Al Hawsawi KA, Alkhudaidi IT, et al. Efficacy and safety of topical clascoterone cream for treatment of acne vulgaris: a systematic review and meta‐analysis of randomized placebo‐controlled trials. <i>Dermatol Ther</i>. 2021;34:e14609. <br/><br/>53. Clasoterone. Package insert. Cassiopea Inc; 2020. <br/><br/>54. Paik J. Topical minocycline foam 4%: a review in acne vulgaris. <i>Am J Clin Dermatol</i>. 2020;21:449-456. <br/><br/>55. Jones TM, Ellman H. Pharmacokinetic comparison of once-daily topical minocycline foam 4% vs oral minocycline for moderate-to-severe acne. <i>J Drugs Dermatol</i>. 2017;16:1022-1028. <br/><br/>56. Minocycline hydrochloride extended-release tablets. Package insert. JG Pharma; July 2020. Accessed January 8, 2024. https://www.jgpharmainc.com/assets/pdf/minocycline-hydrochloride.pdf <br/><br/>57. Dinnendahl V, Fricke U (eds). <i>Arzneistoff-Profile: Basisinformation </i><i>über</i><i> arzneiliche Wirkstoffe</i>. Govi Pharmazeutischer Verlag; 2010. <br/><br/>58. Martins AM, Marto JM, Johnson JL, et al. A review of systemic minocycline side effects and topical minocycline as a safer alternative for treating acne and rosacea. <i>Antibiotics</i>. 2021;10:757. <br/><br/>59. Landis MN. Optimizing isotretinoin treatment of acne: update on current recommendations for monitoring, dosing, safety, adverse effects, compliance, and outcomes. <i>Am J Clin Dermatol</i>. 2020;21:411-419. <br/><br/>60. Draghici C-C, Miulescu R-G, Petca R-C, et al. Teratogenic effect of isotretinoin in both fertile females and males. <i>Exp Ther Med</i>. 2021;21:1-5. <br/><br/>61. Barker RA, Wilcox C, Layton AM. Oral spironolactone for acne vulgaris in adult females: an update of the literature. <i>Am J Clin Dermatol</i>. 2020;21:303-305. <br/><br/>62. Han JJ, Faletsky A, Barbieri JS, et al. New acne therapies and updates on use of spironolactone and isotretinoin: a narrative review. <i>Dermatol Ther (Heidelb)</i>. 2021;11:79-91. <br/><br/>63. Briggs GG, Freeman RK, Yaffe SJ. <i>Drugs in Pregnancy and Lactation: A Reference Guide to Fetal and Neonatal Risk</i>. Lippincott Williams &amp; Wilkins; 2012.<br/><br/>64. Patel DJ, Bhatia N. Oral antibiotics for acne. <i>Am J Clin Dermatol</i>. 2021;22:193-204. <br/><br/>65. Jick H, Holmes LB, Hunter JR, et al. First-trimester drug use and congenital disorders. <i>JAMA</i>. 1981;246:343-346. </p> <p class="reference">66. Valente Duarte de Sousa IC. An overview of sarecycline for the treatment of moderate-to-severe acne vulgaris. <i>Exp Opin Pharmacother</i>. 2021;22:145-154. <br/><br/>67. Hussar DA, Chahine EB. Omadacycline tosylate, sarecycline hydrochloride, rifamycin sodium, and moxidectin. <i>J Am Pharm Assoc</i>. 2019;59:756-760. <br/><br/>68. Haidari W, Bruinsma R, Cardenas-de la Garza JA, et al. Sarecycline review. <i>Ann Pharmacother</i>. 2020;54:164-170. <br/><br/>69. Feldman S, Careccia RE, Barham KL, et al. Diagnosis and treatment of acne. <i>Am Fam Physician</i>. 2004;69:2123-2130. <br/><br/>70. Gammon WR, Meyer C, Lantis S, et al. Comparative efficacy of oral erythromycin versus oral tetracycline in the treatment of acne vulgaris: a double-blind study. <i>J Am Acad Dermatol</i>. 1986;14:183-186. <br/><br/>71. Källén BA, Olausson PO, Danielsson BR. Is erythromycin therapy teratogenic in humans? <i>Reprod Toxicol</i>. 2005;20:209-214. <br/><br/>72. McCormack WM, George H, Donner A, et al. Hepatotoxicity of erythromycin estolate during pregnancy. <i>Antimicrob Agents Chemother</i>. 1977;12:630-635. <br/><br/>73. Cervantes J, Eber AE, Perper M, et al. The role of zinc in the treatment of acne: a review of the literature. <i>Dermatolog Ther</i>. 2018;31:e12576. <br/><br/>74. Dréno B, Blouin E. Acne, pregnant women and zinc salts: a literature review [in French]. <i>Ann Dermatol Venereol.</i> 2008;135:27-33.<br/><br/>75. Eid MM, Saleh MS, Allam NM, et al. Narrow band ultraviolet B versus red light-emitting diodes in the treatment of facial acne vulgaris: a randomized controlled trial. <i>Photobiomodul Photomed Laser Surg</i>. 2021;39:418-424. <br/><br/>76. Zeichner JA. Narrowband UV-B phototherapy for the treatment of acne vulgaris during pregnancy. <i>Arch Dermatol</i>. 2011;147:537-539. <br/><br/>77. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. <i>Lasers Med Sci</i>. 2011;26:481-485. <br/><br/>78. Park KK, Murase JE. Narrowband UV-B phototherapy during pregnancy and folic acid depletion. <i>Arch Dermatol</i>. 2012;148:132-133. <br/><br/>79. Jablonski NG. A possible link between neural tube defects and ultraviolet light exposure. <i>Med Hypotheses</i>. 1999;52:581-582. <br/><br/>80. Zhang M, Goyert G, Lim HW. Folate and phototherapy: what should we inform our patients? <i>J Am Acad Dermatol</i>. 2017;77:958-964. <br/><br/>81. AviClear. Cutera website. Accessed January 8, 2024. https://www.cutera.com/solutions/aviclear/<br/><br/>82. Wu X, Yang Y, Wang Y, et al. Treatment of refractory acne using selective sebaceous gland electro-thermolysis combined with non-thermal plasma. <i>J Cosmet Laser Ther</i>. 2021;23:188-194. <br/><br/>83. Ahn GR, Kim JM, Park SJ, et al. Selective sebaceous gland electrothermolysis using a single microneedle radiofrequency device for acne patients: a prospective randomized controlled study. <i>Lasers Surg Med</i>. 2020;52:396-401. <br/><br/>84. Fabbrocini G, De Padova MP, Tosti A. Chemical peels: what’s new and what isn’t new but still works well. <i>Facial Plast Surg</i>. 2009;25:329-336. <br/><br/>85. Andersen FA. Final report on the safety assessment of glycolic acid, ammonium, calcium, potassium, and sodium glycolates, methyl, ethyl, propyl, and butyl glycolates, and lactic acid, ammonium, calcium, potassium, sodium, and TEA-lactates, methyl, ethyl, isopropyl, and butyl lactates, and lauryl, myristyl, and cetyl lactates. <i>Int J Toxicol</i>. 1998;17(1_suppl):1-241. <br/><br/>86. Lee KC, Korgavkar K, Dufresne RG Jr, et al. Safety of cosmetic dermatologic procedures during pregnancy. <i>Dermatol Surg</i>. 2013;39:1573-1586. <br/><br/>87. James AH, Brancazio LR, Price T. Aspirin and reproductive outcomes. <i>Obstet Gynecol Surv</i>. 2008;63:49-57. <br/><br/>88. Zhou W-S, Xu L, Xie S-H, et al. Decreased birth weight in relation to maternal urinary trichloroacetic acid levels. <i>Sci Total Environ</i>. 2012;416:105-110. <br/><br/>89. Schwartz DB, Greenberg MD, Daoud Y, et al. Genital condylomas in pregnancy: use of trichloroacetic acid and laser therapy. <i>Am J Obstet Gynecol</i>. 1988;158:1407-1416. <br/><br/>90. Starkman SJ, Mangat DS. Chemical peel (deep, medium, light). <i>Facial Plast Surg Clin North Am</i>. 2020;28:45-57. <br/><br/>91. Trivedi M, Kroumpouzos G, Murase J. A review of the safety of cosmetic procedures during pregnancy and lactation. <i>Int J Womens Dermatol</i>. 2017;3:6-10. <br/><br/>92. Gallagher T, Taliercio M, Nia JK, et al. Dermatologist use of intralesional triamcinolone in the treatment of acne. <i>J Clin Aesthet Dermatol</i>. 2020;13:41-43. <br/><br/>93. Zamil DH, Burns EK, Perez-Sanchez A, et al. Risk of birth defects from vitamin A “acne supplements” sold online. <i>Dermatol Pract Concept</i>. 2021;11:e2021075.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>The management of acne in pregnancy requires careful consideration of therapeutic choices to guarantee the safety of both the mother and the developing fetus.</li> <li>The use of topicals should be observed as first-line therapy, but consideration for systemic therapy in cases of treatment failure or more severe disease is warranted.</li> <li>Discussion of patient expectations and involving them in decision-making for therapeutic choice is crucial.</li> </ul> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Drs. Yaghi and Keri are from the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Florida. Dr. Keri also is from Dermatology Service, Miami VA Hospital, Florida. Daniela Baboun is from Herbert Wertheim College of Medicine, Florida International University, Miami.</p> <p class="disclosure">Dr. Yaghi and Daniela Baboun report no conflict of interest. Dr. Keri is on the advisory board for Ortho Dermatologics, has received research funding from Galderma, and has received honoraria from Merck Manuals. <br/><br/>Correspondence: Jonette E. Keri, MD, PhD, University of Miami Miller School of Medicine, 1600 NW 10th Ave, RSMB Room 2023A, Miami, FL 33136 (jkeri@med.miami.edu).<br/><br/>doi:10.12788/cutis.0951</p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • The management of acne in pregnancy requires careful consideration of therapeutic choices to guarantee the safety of both the mother and the developing fetus.
  • The use of topicals should be observed as first-line therapy, but consideration for systemic therapy in cases of treatment failure or more severe disease is warranted.
  • Discussion of patient expectations and involving them in decision-making for therapeutic choice is crucial.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026AC.SIG
Disable zoom
Off

Preventing ASCVD Events: Using Coronary Artery Calcification Scores to Personalize Risk and Guide Statin Therapy

Article Type
Changed
Fri, 01/12/2024 - 10:13

 

Lung cancer is the most common cause of cancer mortality, and cigarette smoking is the most significant risk factor. Several randomized clinical trials have shown that lung cancer screening (LCS) with nonelectrocardiogram (ECG)-gated low-dose computed tomography (LDCT) reduces both lung cancer and all-cause mortality.1,2 Hence, the US Preventive Screening Task Force (USPSTF) recommends annual screening with LDCT in adults aged 50 to 80 years who have a 20-pack-year smoking history and currently smoke or have quit within the past 15 years.3

Smoking is also an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), and LCS clinical trials acknowledge that mortality from ASCVD events exceeds that of lung cancer.4,5 In an analysis of asymptomatic individuals from the Framingham Heart Offspring study who were eligible for LCS, the ASCVD event rate during a median (IQR) follow-up of 11.4 (9.7-12.0) years was 12.6%.6 However, despite the high rate of ASCVD events in this population, primary prevention strategies are consistently underused. In a study of 5495 individuals who underwent LCS with LDCT, only 40% of those eligible for statins had one prescribed, underscoring the missed opportunity for preventing ASCVD events during LCS.7 Yet the interactions for shared decision making and the availability of coronary artery calcification (CAC) scores from the LDCT provide an ideal window for intervening and preventing ASCVD events during LCS.

CAC is a hallmark of atherosclerotic plaque development and is proportional to plaque burden and ASCVD risk.8 Because of the relationship between CAC, subclinical atherosclerosis, and ASCVD risk, there is an opportunity to use CAC detected by LDCT to predict ASCVD risk and guide recommendations for statin treatment in individuals enrolled in LCS. Traditionally, CAC has been visualized by ECG-gated noncontrast CT scans with imaging protocols specifically designed to visualize the coronary arteries, minimize motion artifacts, and reduce signal noise. These scans are specifically done for primary prevention risk assessment and report an Agatston score, a summed measure based on calcified plaque area and maximal density.9 Results are reported as an overall CAC score and an age-, sex-, and race-adjusted percentile of CAC. Currently, a CAC score ≥ 100 or above the 75th percentile for age, sex, and race is considered abnormal.

High-quality evidence supports CAC scores as a strong predictor of ASCVD risk independent of age, sex, race, and other traditional risk factors.10-12 In asymptomatic individuals, a CAC score of 0 is a strong, negative risk factor associated with very low annualized mortality rates and cardiovascular (CV) events, so intermediate-risk individuals can be reclassified to a lower risk group avoiding or delaying statin therapy.13 As a result, current primary prevention guidelines allow for CAC scoring in asymptomatic, intermediate-risk adults where the clinical benefits of statin therapy are uncertain, knowing the CAC score will aid in the clinical decision to delay or initiate statin therapy.

Unlike traditional ECG-gated CAC scoring, LDCT imaging protocols are non–ECG-gated and performed at variable energy and slice thickness to optimize the detection of lung nodules. Early studies suggested that CAC detected by LDCT could be used in lieu of traditional CAC scoring to personalize risk.14,15 Recently, multiple studies have validated the accuracy and reproducibility of LDCT to detect and quantify CAC. In both the NELSON and the National Lung Screening Trial (NLST) LCS trials, higher visual and quantitative measures of CAC were independently and incrementally associated with ASCVD risk.16,17 A subsequent review and meta-analysis of 6 LCS trials confirmed CAC detected by LDCT to be an independent predictor of ASCVD events regardless of the method used to measure CAC.18

table.png

There is now consensus that either an Agatston score or a visual estimate of CAC be reported on all noncontrast, noncardiac chest CT scans irrespective of the indication or technique, including LDCT scans for LCS using a uniform reporting system known as the Coronary Artery Calcium Data and Reporting System (CAC-DRS).19 The CAC-DRS simplifies reporting and adds modifiers indicating if the reported score is visual (V) or Agatston (A) and number of vessels involved. For example, CAC-DRS A0 or CAC-DRS V0 would indicate an Agatston score of 0 or a visual score of 0. CAC-DRS A1/N2 would indicate a total Agatston score of 1-99 in 2 coronary arteries. The currently agreed-on CAC-DRS risk groups are listed in the Table, along with their corresponding visual score or Agatston score and anticipated 10-year event rate, irrespective of other risk factors.20

As LCS efforts increase, primary care practitioners will receive LDCT reports that now incorporate an estimation of CAC (visual or quantitative). Thus, it will be increasingly important to know how to interpret and use these scores to guide clinical decisions regarding the initiation of statin therapy, referral for additional testing, and when to seek specialty cardiology care. For instance, does the absence of CAC (CAC = 0) on LDCT predict a low enough risk for statin therapy to be delayed or withdrawn? Does increasing CAC scores on follow-up LDCT in individuals on statin therapy represent treatment failure? When should CAC scores trigger additional testing, such as a stress test or referral to cardiology specialty care?

 

 

Primary Prevention in LCS

The initial approach to primary prevention in LCS is no different from that recommended by the 2018 multisociety guidelines on the management of blood cholesterol, the 2019 American College of Cardiology/American Heart Association (ACC/AHA) guideline on primary prevention, or the 2022 USPTSF recommendations on statin use for primary prevention of CV disease in adults.21-23 For a baseline low-density lipoprotein cholesterol (LDL-C) ≥ 190 mg/dL, high-intensity statin therapy is recommended without further risk stratification. Individuals with diabetes also are at higher-than-average risk, and moderate-intensity statin therapy is recommended.

For individuals not in either group, a validated ASCVD risk assessment tool is recommended to estimate baseline risk. The most validated tool for estimating risk in the US population is the 2013 ACC/AHA Pooled Cohort Equation (PCE) which provides an estimate of the 10-year risk for fatal and myocardial infarction and fatal and nonfatal stroke.24 The PCE risk calculator uses age, presence of diabetes, sex, smoking history, total cholesterol, high-density lipoprotein cholesterol, systolic blood pressure, and treatment for hypertension to place individuals into 1 of 4 risk groups: low (< 5%), borderline (5% to < 7.5%), intermediate (≥ 7.5% to < 20%), and high (≥ 20%). Clinicians should be aware that the PCE only considers current smoking history and not prior smoking history or cumulative pack-year history. This differs from eligibility for LCS where recent smoking plays a larger role. All these risk factors are important to consider when evaluating risk and discussing risk-reducing strategies like statin therapy.

The 2018 multisociety guidelines and the 2019 primary prevention guidelines set the threshold for considering initiation of statin therapy at intermediate risk ≥ 7.5%.21,22 The 2020 US Department of Veterans Affairs/Department of Defense guidelines set the threshold for considering statin therapy at an estimated 10-year event rate of 12%, whereas the 2022 UPSTF recommendations set the threshold at 10% with additional risk factors as the threshold for statin therapy.23,25 The reasons for these differences are beyond the scope of this review, but all these guidelines use the PCE to estimate baseline risk as the starting point for clinical decision making.

The PCE was originally derived and validated in population studies dating to the 1960s when the importance of diet, exercise, and smoking cessation in reducing ASCVD events was not well appreciated. The application of the PCE in more contemporary populations shows that it overestimates risk, especially in older individuals and women.26,27 Overestimation of risk has the potential to result in the initiation of statin therapy in individuals in whom the actual clinical benefit would otherwise be small.

figure.png

To address this issue, current guidelines allow the use of CAC scoring to refine risk in individuals who are classified as intermediate risk and who otherwise desire to avoid lifelong statin therapy. Using current recommendations, we make suggestions on how to use CAC scores from LDCT to aid in clinical decision making for individuals in LCS (Figure).

No Coronary Artery Calcification

Between 25% and 30% of LDCT done for LCS will show no CAC.14,16 In general population studies, a CAC score of 0 is a strong negative predictor when there are no other risk factors.13,28 In contrast, the negative predictive ability of a CAC score of 0 in individuals with a smoking history who are eligible for LCS is unproven. In multivariate modeling, a CAC score of 0 did not reduce the significant hazard of all-cause mortality in patients with diabetes or smokers.29 In an analysis of 44,042 individuals without known heart disease referred for CAC scoring, the frequency of a CAC score of 0 was only modestly lower in smokers (38%) compared with nonsmokers (42%), yet the all-cause mortality rate was significantly higher.30 In addition, Multi-Ethnic Study of Atherosclerosis (MESA) participants who were current smokers or eligible for LCS and had a CAC score of 0 had an observed 11-year ASCVD event rate of 13.4% and 20.8%, respectively, leading to the conclusion that a CAC score of 0 may not be predictive of minimal risk in smokers and those eligible for LCS.31 Additionally, in LCS-eligible individuals, the PCE underestimated event rates and incorporation of CAC scores did not significantly improve risk estimation. Finally, data from the NLST screening trial showed that the absence of CAC on LDCT was not associated with better survival or lower CV mortality compared with individuals with low CAC scores.32

 

 

The question of whether individuals undergoing LCS with LDCT who have no detectable CAC can avoid statin therapy is an unresolved issue; no contemporary studies have looked specifically at the relationship between estimated risk, a CAC score of 0, and ASCVD outcomes in individuals participating in LCS. For these reasons, we recommend moderate-intensity statin therapy when the estimated risk is intermediate because it is unclear that either an Agatston score of 0 reclassifies intermediate-risk LCS-eligible individuals to a lower risk group.

For the few borderline risk (estimated risk, 5% to < 7.5%) LCS-eligible individuals, a CAC score of 0 might confer low short-term risk but the long-term benefit of statin therapy on reducing subsequent risk, the presence of other risk factors, and the willingness to stop smoking should all be considered. For these individuals who elect to avoid statin therapy, annual re-estimation of risk at the time of repeat LDCT is recommended. In these circumstances, referral for traditional Agatston scoring is not likely to change decision making because the sensitivity of the 2 techniques is very similar.

Agatston Score of 1-99 or CAC-DRS or Visual Score of 1

In general population studies, these scores correspond to borderline risk and an estimated 10-year event rate of just under 7.5%.20 In both the NELSON and NLST LCS trials, even low amounts of CAC regardless of the scoring method were associated with higher observed ASCVD mortality when adjusted for other baseline risk factors.32 Thus, in patients undergoing LCS with intermediate and borderline risk, a CAC score between 1 and 99 or a visual estimate of 1 indicates the presence of subclinical atherosclerosis, and moderate-intensity statin therapy is reasonable.

Agatston Score of 100-299 or CAC-DRS or Visual Score of 2

Across all ages, races, and sexes, CAC scores between 100 to 299 are associated with an event rate of about 15% over 10 years.20 In the NELSON LCS trial, the adjusted hazard ratio for ASCVD events with a nontraditional Agatston score of 101 to 400 was 6.58.33 Thus, in patients undergoing LCS with a CAC score of 100 to 299, regardless of the baseline risk estimate, the projected absolute event rate at 10 years would be about 20%. Moderate-intensity statin therapy is recommended to reduce the baseline LDL-C by 30% to 49%.

Agatston Score of > 300 orCAC-DRS or Visual Score of 3

Agatston CAC scores > 300 are consistent with a 10-year incidence of ASCVD events of > 15% regardless of age, sex, or race and ethnicity.20 In the Calcium Consortium, a CAC > 400 was correlated with an event rate of 13.6 events/1000 person-years.12 In a Walter Reed Military Medical Center study, a CAC score > 400 projected a cumulative incidence of ASCVD events of nearly 20% at 10 years.34 In smokers eligible for LCS, a CAC score > 300 projected a 10-year ASCVD event rate of 25%.29 In these patients, moderate-intensity statin therapy is recommended, although high-intensity statin therapy can be considered if there are other risk factors.

Agatston Score ≥ 1000

The 2018 consensus statement on CAC reporting categorizes all CAC scores > 300 into a single risk group because the recommended treatment options do not differ.19 However, recent data suggest this might not be the case since individuals with very high CAC scores experience high rates of events that might justify more aggressive intervention. In an analysis of individuals who participated in the CAC Consortium with a CAC score ≥ 1000, the all-cause mortality rate was 18.8 per 1000 person-years with a CV mortality rate of 8 per 1000 person-years.35 Individuals with very high levels of CAC > 1000 also have a greater number of diseased coronary arteries, higher involvement of the left main coronary artery, and significantly higher event rates compared with those with a CAC of 400 to 999.36 In an analysis of individuals from the NLST trial, nontraditionally measured Agatston score > 1000 was associated with a hazard ratio for coronary artery disease (CAD) mortality of 3.66 in men and 5.81 in women.17 These observed and projected levels of risk are like that seen in secondary prevention trials, and some experts have recommended the use of high-intensity statin therapy to reduce LDL-C to < 70 mg/dL.37

Primary Prevention in Individuals aged 76 to 80 years

LCS can continue through age 80 years, while the PCE and primary prevention guidelines are truncated at age 75 years. Because age is a major contributor to risk, many of these individuals will already be in the intermediate- to high-risk group. However, the net clinical benefit of statin therapy for primary prevention in this age group is not well established, and the few primary prevention trials in this group have not demonstrated net clinical benefit.38 As a result, current guidelines do not provide specific treatment recommendations for individuals aged > 75 years but recognize the value of shared decision making considering associated comorbidities, age-related risks of statin therapy, and the desires of the individual to avoid ASCVD-related events even if the net clinical benefit is low.

Older individuals with elevated CAC scores should be informed about the risk of ASCVD events and the potential but unproven benefit of moderate-intensity statin therapy. Older individuals with a CAC score of 0 likely have low short-term risk of ASCVD events and withholding statin therapy is not unreasonable.

 

 

CAC Scores on Annual LDCT Scans

Because LCS requires annual LDCT scans, primary care practitioners and patients need to understand the significance of changing CAC scores over time. For individuals not on statin therapy, increasing calcification is a marker of progression of subclinical atherosclerosis. Patients undergoing LCS not on statin who have progressive increases in their CAC should consider initiating statin therapy. Individuals who opted not to initiate statin therapy who subsequently develop CAC should be re-engaged in a discussion about the significance of the finding and the clinically proven benefits of statin therapy in individuals with subclinical atherosclerosis. These considerations do not apply to individuals already on statin therapy. Statins convert lipid-rich plaques to lipid-depleted plaques, resulting in increasing calcification. As a result, CAC scores do not decrease and may increase with statin therapy.39 Individuals participating in annual LCS should be informed of this possibility. Also, in these individuals, referral to specialty care as a treatment failure is not supported by the literature.

Furthermore, serial CAC scoring to titrate the intensity of statin therapy is not currently recommended. The goal with moderate-intensity statin therapy is a 30% to 49% reduction from baseline LDL-C. If this milestone is not achieved, the statin dose can be escalated. For high-intensity statin therapy, the goal is a > 50% reduction. If this milestone is not achieved, then additional lipid-lowering agents, such as ezetimibe, can be added.

Further ASCVD Testing

LCS with LDCT is associated with improved health outcomes, and LDCT is the preferred imaging modality. The ability of LDCT to detect and quantify CAC is sufficient for clinical decision making. Therefore, obtaining a traditional CAC score increases radiation exposure without additional clinical benefits.

Furthermore, although referral for additional testing in those with nonzero CAC scores is common, current evidence does not support this practice in asymptomatic individuals. Indeed, the risks of LCS include overdiagnosis, excessive testing, and overtreatment secondary to the discovery of other findings, such as benign pulmonary nodules and CAC. With respect to CAD, randomized controlled trials do not support a strategy of coronary angiography and intervention in asymptomatic individuals, even with moderate-to-severe ischemia on functional testing.40 As a result, routine stress tests to diagnose CAD or to confirm the results of CAC scores in asymptomatic individuals are not recommended. The only potential exception would be in select cases where the CAC score is > 1000 and when calcium is predominately located in the left main coronary artery.

Conclusions

LCS provides smokers at risk for lung cancer with the best probability to survive that diagnosis, and coincidentally LCS may also provide the best opportunity to prevent ASCVD events and mortality. Before initiating LCS, clinicians should initiate a shared decision making conversation about the benefits and risks of LDCT scans. In addition to relevant education about smoking, during shared decision making, the initial ASCVD risk estimate should be done using the PCE and when appropriate the benefits of statin therapy discussed. Individuals also should be informed of the potential for identifying CAC and counseled on its significance and how it might influence the decision to recommend statin therapy.

In patients undergoing LCS with an estimated risk of ≥ 7.5% to < 20%, moderate-intensity statin therapy is indicated. In this setting, a CAC score > 0 indicates subclinical atherosclerosis and should be used to help direct patients toward initiating statin therapy. Unfortunately, in patients undergoing LCS a CAC score of 0 might not provide protection against ASCVD, and until there is more information to the contrary, these individuals should at least participate in shared decision making about the long-term benefits of statin therapy in reducing ASCVD risk. Because LDCT scanning is done annually, there are opportunities to review the importance of prevention and to adjust therapy as needed to achieve the greatest reduction in ASCVD. Reported elevated CAC scores on LDCT provide an opportunity to re-engage the patient in the discussion about the benefits of statin therapy if they are not already on a statin, or consideration for high-intensity statin if the CAC score is > 1000 or reduction in baseline LDL-C is < 30% on the current statin dose.

References

1. de Koning HJ, van der Aalst CM, Oudkerk M. Lung-cancer screening and the NELSON Trial. Reply. N Engl J Med. 2020;382(22):2165-2166. doi:10.1056/NEJMc2004224

2. Aberle T, Adams DR, Berg AM, et al; National Lung Screening Trial Research Team. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):396-409. doi:10.1056/NEJMoa1102873

3. Krist AH, Davidson KW, Mangione CM, et al; US Preventive Services Task Force. Screening for lung cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;25(10):962-970. doi:10.1001/jama.2021.1117

4. Jha P, Ramasundarahettige C, Landsman V. 21st-century hazards of smoking and benefits of cessation in the United States. N Engl J Med. 2013;368(4):341-350. doi:10.1056/NEJMsa1211128

5. Khan SS, Ning H, Sinha A, et al. Cigarette smoking and competing risks for fatal and nonfatal cardiovascular disease subtypes across the life course. J Am Heart Assoc. 2021;10(23):e021751. doi:10.1161/JAHA.121.021751

6. Lu MT, Onuma OK, Massaro JM, et al. Lung cancer screening eligibility in the community: cardiovascular risk factors, coronary artery calcification, and cardiovascular events. Circulation. 2016;134(12):897-899. doi:10.1161/CIRCULATIONAHA.116.023957

7. Tailor TD, Chiles C, Yeboah J, et al. Cardiovascular risk in the lung cancer screening population: a multicenter study evaluating the association between coronary artery calcification and preventive statin prescription. J Am Coll Radiol. 2021;18(9):1258-1266. doi:10.1016/j.jacr.2021.01.015

8. Mori H, Torii S, Kutyna M, et al. Coronary artery calcification and its progression: what does it really mean? JACC Cardiovasc Imaging. 2018;11(1):127-142. doi:10.1016/j.jcmg.2017.10.012

10. Nasir K, Bittencourt MS, Blaha MJ, et al. Implications of coronary artery calcium testing among statin candidates according to American College of Cardiology/American Heart Association cholesterol management guidelines: MESA (Multi-Ethnic Study of Atherosclerosis). J Am Coll Cardiol. 2015;66(15): 1657-1668. doi:10.1016/j.jacc.2015.07.066

11. Detrano R, Guerci AD, Carr JJ, et al. Coronary calcium as a predictor of coronary events in four racial or ethnic groups. N Engl J Med. 2008;358(13):1336-1345. doi:10.1056/NEJMoa072100

12. Grandhi GR, Mirbolouk M, Dardari ZA. Interplay of coronary artery calcium and risk factors for predicting CVD/CHD Mortality: the CAC Consortium. JACC Cardiovasc Imaging. 2020;13(5):1175-1186. doi:10.1016/j.jcmg.2019.08.024

13. Blaha M, Budoff MJ, Shaw J. Absence of coronary artery calcification and all-cause mortality. JACC Cardiovasc Imaging. 2009;2(6):692-700. doi:10.1016/j.jcmg.2009.03.009

14. Shemesh J, Henschke CI, Farooqi A, et al. Frequency of coronary artery calcification on low-dose computed tomography screening for lung cancer. Clin Imaging. 2006;30(3):181-185. doi:10.1016/j.clinimag.2005.11.002

15. Shemesh J, Henschke C, Shaham D, et al. Ordinal scoring of coronary artery calcifications on low-dose CT scans of the chest is predictive of death from cardiovascular disease. Radiology. 2010;257:541-548. doi:10.1148/radiol.10100383

16. Jacobs PC, Gondrie MJ, van der Graaf Y, et al. Coronary artery calcium can predict all-cause mortality and cardiovascular events on low-dose CT screening for lung cancer. AJR Am J Roentgenol. 2012;198(3):505-511. doi:10.2214/AJR.10.5577

17. Lessmann N, de Jong PA, Celeng C, et al. Sex differences in coronary artery and thoracic aorta calcification and their association with cardiovascular mortality in heavy smokers. JACC Cardiovasc Imaging. 2019;12(9):1808-1817. doi:10.1016/j.jcmg.2018.10.026

18. Gendarme S, Goussault H, Assie JB, et al. Impact on all-cause and cardiovascular mortality rates of coronary artery calcifications detected during organized, low-dose, computed-tomography screening for lung cancer: systematic literature review and meta-analysis. Cancers (Basel). 2021;13(7):1553. doi:10.3390/cancers13071553

19. Hecht HS, Blaha MJ, Kazerooni EA, et al. CAC-DRS: coronary artery calcium data and reporting system. An expert consensus document of the Society of Cardiovascular Computed Tomography (SCCT). J Cardiovasc Comput Tomogr. 2018;12(3):185-191. doi:10.1016/j.jcct.2018.03.008

20. Budoff MJ, Young R, Burke G, et al. Ten-year association of coronary artery calcium with atherosclerotic cardiovascular disease (ASCVD) events: the multi-ethnic study of atherosclerosis (MESA). Eur Heart J. 2018;39(25):2401-2408. doi:10.1093/eurheartj/ehy217

21. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139(25):e1046-e1081. doi:10.1161/CIR.0000000000000624

22. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678

23. Mangione CM, Barry MJ, Nicholson WK, et al; US Preventive Services Task Force. Statin use for the primary prevention of cardiovascular disease in adults: US Preventive Services Task Force recommendation statement. JAMA. 2022;328(8):746-753. doi:10.1001/jama.2022.13044

24. Stone NJ, Robinson JG, Lichtenstein AH, et al; American College of Cardiology/American Heart Association Task Force on Practice. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63(25 pt B):2889-2934. doi:10.1016/j.jacc.2013.11.002

<--pagebreak-->

25. US Department of Veterans Affairs, Department of Defense. VA/DoD clinical practice guideline. Updated August 25, 2021. Accessed November 3, 2023. https://www.healthquality.va.gov/guidelines/cd/lipids

26. DeFilippis AP, Young, R, Carrubba CJ, et al. An analysis of calibration and discrimination among multiple cardiovascular risk scores in a modern multiethnic cohort. Ann Intern Med. 2015;162(4):266-275. doi:10.7326/M14-1281

27. Rana JS, Tabada GH, Solomon, MD, et al. Accuracy of the atherosclerotic cardiovascular risk equation in a large contemporary, multiethnic population. J Am Coll Cardiol. 2016;67(18):2118-2130. doi:10.1016/j.jacc.2016.02.055

28. Sarwar A, Shaw LJ, Shapiro MD, et al. Diagnostic and prognostic value of absence of coronary artery calcification. JACC Cardiovasc Imaging. 2009;2(6):675-688. doi:10.1016/j.jcmg.2008.12.031

29. McEvoy JW, Blaha MJ, Rivera JJ, et al. Mortality rates in smokers and nonsmokers in the presence or absence of coronary artery calcification. JACC Cardiovasc Imaging. 2012;5(10):1037-1045. doi:10.1016/j.jcmg.2012.02.017

30. Leigh A, McEvoy JW, Garg P, et al. Coronary artery calcium scores and atherosclerotic cardiovascular disease risk stratification in smokers. JACC Cardiovasc Imaging. 2019;12(5):852-861. doi:10.1016/j.jcmg.2017.12.017

31. Garg PK, Jorgensen NW, McClelland RL, et al. Use of coronary artery calcium testing to improve coronary heart disease risk assessment in lung cancer screening population: The Multi-Ethnic Study of Atherosclerosis (MESA). J Cardiovasc Comput Tomagr. 2018;12(6):439-400.

32. Chiles C, Duan F, Gladish GW, et al. Association of coronary artery calcification and mortality in the national lung screening trial: a comparison of three scoring methods. Radiology. 2015;276(1):82-90. doi:10.1148/radiol.15142062

33. Takx RA, Isgum I, Willemink MJ, et al. Quantification of coronary artery calcium in nongated CT to predict cardiovascular events in male lung cancer screening participants: results of the NELSON study. J Cardiovasc Comput Tomogr. 2015;9(1):50-57. doi:10.1016/j.jcct.2014.11.006

34. Mitchell JD, Paisley R, Moon P, et al. Coronary artery calcium and long-term risk of death, myocardial infarction, and stroke: The Walter Reed Cohort Study. JACC Cardiovasc Imaging. 2018;11(12):1799-1806. doi:10.1016/j.jcmg.2017.09.003

35. Peng AW, Mirbolouk M, Orimoloye OA, et al. Long-term all-cause and cause-specific mortality in asymptomatic patients with CAC >/=1,000: results from the CAC Consortium. JACC Cardiovasc Imaging. 2019;13(1, pt 1):83-93. doi:10.1016/j.jcmg.2019.02.005

36. Peng AW, Dardari ZA. Blumenthal RS, et al. Very high coronary artery calcium (>/=1000) and association with cardiovascular disease events, non-cardiovascular disease outcomes, and mortality: results from MESA. Circulation. 2021;143(16):1571-1583. doi:10.1161/CIRCULATIONAHA.120.050545

37. Orringer CE, Blaha MJ, Blankstein R, et al. The National Lipid Association scientific statement on coronary artery calcium scoring to guide preventive strategies for ASCVD risk reduction. J Clin Lipidol. 2021;15(1):33-60. doi:10.1016/j.jacl.2020.12.005

38. Sheperd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease. (PROSPER): a randomized controlled trial. Lancet. 2002;360:1623-1630. doi:10.1016/s0140-6736(02)11600-x

39. Puri R, Nicholls SJ, Shao M, et al. Impact of statins on serial coronary calcification during atheroma progression and regression. J Am Coll Cardiol. 2015;65(13):1273-1282. doi:10.1016/j.jacc.2015.01.036

40. Maron D.J, Hochman J S, Reynolds HR, et al; ISCHEMIA Research Group. Initial invasive or conservative strategy for stable coronary disease. N Engl J Med. 2020;382(15):1395-1407. doi:10.1056/NEJMoa1915922

Article PDF
Author and Disclosure Information

Joel Kupfer, MDa; Helme Silvet, MDb; Samuel M. Aguayo, MDa

Correspondence:  Joel Kupfer  (joel.kupfer@va.gov)

aCarl T. Hayden Veterans Affairs Medical Center, Phoenix, Arizona

bVeterans Affairs Loma Linda Healthcare System, California

Author disclosures
The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies.

Issue
Federal Practitioner - 41(1)a
Publications
Topics
Page Number
1
Sections
Author and Disclosure Information

Joel Kupfer, MDa; Helme Silvet, MDb; Samuel M. Aguayo, MDa

Correspondence:  Joel Kupfer  (joel.kupfer@va.gov)

aCarl T. Hayden Veterans Affairs Medical Center, Phoenix, Arizona

bVeterans Affairs Loma Linda Healthcare System, California

Author disclosures
The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies.

Author and Disclosure Information

Joel Kupfer, MDa; Helme Silvet, MDb; Samuel M. Aguayo, MDa

Correspondence:  Joel Kupfer  (joel.kupfer@va.gov)

aCarl T. Hayden Veterans Affairs Medical Center, Phoenix, Arizona

bVeterans Affairs Loma Linda Healthcare System, California

Author disclosures
The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the U.S. Government, or any of its agencies.

Article PDF
Article PDF

 

Lung cancer is the most common cause of cancer mortality, and cigarette smoking is the most significant risk factor. Several randomized clinical trials have shown that lung cancer screening (LCS) with nonelectrocardiogram (ECG)-gated low-dose computed tomography (LDCT) reduces both lung cancer and all-cause mortality.1,2 Hence, the US Preventive Screening Task Force (USPSTF) recommends annual screening with LDCT in adults aged 50 to 80 years who have a 20-pack-year smoking history and currently smoke or have quit within the past 15 years.3

Smoking is also an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), and LCS clinical trials acknowledge that mortality from ASCVD events exceeds that of lung cancer.4,5 In an analysis of asymptomatic individuals from the Framingham Heart Offspring study who were eligible for LCS, the ASCVD event rate during a median (IQR) follow-up of 11.4 (9.7-12.0) years was 12.6%.6 However, despite the high rate of ASCVD events in this population, primary prevention strategies are consistently underused. In a study of 5495 individuals who underwent LCS with LDCT, only 40% of those eligible for statins had one prescribed, underscoring the missed opportunity for preventing ASCVD events during LCS.7 Yet the interactions for shared decision making and the availability of coronary artery calcification (CAC) scores from the LDCT provide an ideal window for intervening and preventing ASCVD events during LCS.

CAC is a hallmark of atherosclerotic plaque development and is proportional to plaque burden and ASCVD risk.8 Because of the relationship between CAC, subclinical atherosclerosis, and ASCVD risk, there is an opportunity to use CAC detected by LDCT to predict ASCVD risk and guide recommendations for statin treatment in individuals enrolled in LCS. Traditionally, CAC has been visualized by ECG-gated noncontrast CT scans with imaging protocols specifically designed to visualize the coronary arteries, minimize motion artifacts, and reduce signal noise. These scans are specifically done for primary prevention risk assessment and report an Agatston score, a summed measure based on calcified plaque area and maximal density.9 Results are reported as an overall CAC score and an age-, sex-, and race-adjusted percentile of CAC. Currently, a CAC score ≥ 100 or above the 75th percentile for age, sex, and race is considered abnormal.

High-quality evidence supports CAC scores as a strong predictor of ASCVD risk independent of age, sex, race, and other traditional risk factors.10-12 In asymptomatic individuals, a CAC score of 0 is a strong, negative risk factor associated with very low annualized mortality rates and cardiovascular (CV) events, so intermediate-risk individuals can be reclassified to a lower risk group avoiding or delaying statin therapy.13 As a result, current primary prevention guidelines allow for CAC scoring in asymptomatic, intermediate-risk adults where the clinical benefits of statin therapy are uncertain, knowing the CAC score will aid in the clinical decision to delay or initiate statin therapy.

Unlike traditional ECG-gated CAC scoring, LDCT imaging protocols are non–ECG-gated and performed at variable energy and slice thickness to optimize the detection of lung nodules. Early studies suggested that CAC detected by LDCT could be used in lieu of traditional CAC scoring to personalize risk.14,15 Recently, multiple studies have validated the accuracy and reproducibility of LDCT to detect and quantify CAC. In both the NELSON and the National Lung Screening Trial (NLST) LCS trials, higher visual and quantitative measures of CAC were independently and incrementally associated with ASCVD risk.16,17 A subsequent review and meta-analysis of 6 LCS trials confirmed CAC detected by LDCT to be an independent predictor of ASCVD events regardless of the method used to measure CAC.18

table.png

There is now consensus that either an Agatston score or a visual estimate of CAC be reported on all noncontrast, noncardiac chest CT scans irrespective of the indication or technique, including LDCT scans for LCS using a uniform reporting system known as the Coronary Artery Calcium Data and Reporting System (CAC-DRS).19 The CAC-DRS simplifies reporting and adds modifiers indicating if the reported score is visual (V) or Agatston (A) and number of vessels involved. For example, CAC-DRS A0 or CAC-DRS V0 would indicate an Agatston score of 0 or a visual score of 0. CAC-DRS A1/N2 would indicate a total Agatston score of 1-99 in 2 coronary arteries. The currently agreed-on CAC-DRS risk groups are listed in the Table, along with their corresponding visual score or Agatston score and anticipated 10-year event rate, irrespective of other risk factors.20

As LCS efforts increase, primary care practitioners will receive LDCT reports that now incorporate an estimation of CAC (visual or quantitative). Thus, it will be increasingly important to know how to interpret and use these scores to guide clinical decisions regarding the initiation of statin therapy, referral for additional testing, and when to seek specialty cardiology care. For instance, does the absence of CAC (CAC = 0) on LDCT predict a low enough risk for statin therapy to be delayed or withdrawn? Does increasing CAC scores on follow-up LDCT in individuals on statin therapy represent treatment failure? When should CAC scores trigger additional testing, such as a stress test or referral to cardiology specialty care?

 

 

Primary Prevention in LCS

The initial approach to primary prevention in LCS is no different from that recommended by the 2018 multisociety guidelines on the management of blood cholesterol, the 2019 American College of Cardiology/American Heart Association (ACC/AHA) guideline on primary prevention, or the 2022 USPTSF recommendations on statin use for primary prevention of CV disease in adults.21-23 For a baseline low-density lipoprotein cholesterol (LDL-C) ≥ 190 mg/dL, high-intensity statin therapy is recommended without further risk stratification. Individuals with diabetes also are at higher-than-average risk, and moderate-intensity statin therapy is recommended.

For individuals not in either group, a validated ASCVD risk assessment tool is recommended to estimate baseline risk. The most validated tool for estimating risk in the US population is the 2013 ACC/AHA Pooled Cohort Equation (PCE) which provides an estimate of the 10-year risk for fatal and myocardial infarction and fatal and nonfatal stroke.24 The PCE risk calculator uses age, presence of diabetes, sex, smoking history, total cholesterol, high-density lipoprotein cholesterol, systolic blood pressure, and treatment for hypertension to place individuals into 1 of 4 risk groups: low (< 5%), borderline (5% to < 7.5%), intermediate (≥ 7.5% to < 20%), and high (≥ 20%). Clinicians should be aware that the PCE only considers current smoking history and not prior smoking history or cumulative pack-year history. This differs from eligibility for LCS where recent smoking plays a larger role. All these risk factors are important to consider when evaluating risk and discussing risk-reducing strategies like statin therapy.

The 2018 multisociety guidelines and the 2019 primary prevention guidelines set the threshold for considering initiation of statin therapy at intermediate risk ≥ 7.5%.21,22 The 2020 US Department of Veterans Affairs/Department of Defense guidelines set the threshold for considering statin therapy at an estimated 10-year event rate of 12%, whereas the 2022 UPSTF recommendations set the threshold at 10% with additional risk factors as the threshold for statin therapy.23,25 The reasons for these differences are beyond the scope of this review, but all these guidelines use the PCE to estimate baseline risk as the starting point for clinical decision making.

The PCE was originally derived and validated in population studies dating to the 1960s when the importance of diet, exercise, and smoking cessation in reducing ASCVD events was not well appreciated. The application of the PCE in more contemporary populations shows that it overestimates risk, especially in older individuals and women.26,27 Overestimation of risk has the potential to result in the initiation of statin therapy in individuals in whom the actual clinical benefit would otherwise be small.

figure.png

To address this issue, current guidelines allow the use of CAC scoring to refine risk in individuals who are classified as intermediate risk and who otherwise desire to avoid lifelong statin therapy. Using current recommendations, we make suggestions on how to use CAC scores from LDCT to aid in clinical decision making for individuals in LCS (Figure).

No Coronary Artery Calcification

Between 25% and 30% of LDCT done for LCS will show no CAC.14,16 In general population studies, a CAC score of 0 is a strong negative predictor when there are no other risk factors.13,28 In contrast, the negative predictive ability of a CAC score of 0 in individuals with a smoking history who are eligible for LCS is unproven. In multivariate modeling, a CAC score of 0 did not reduce the significant hazard of all-cause mortality in patients with diabetes or smokers.29 In an analysis of 44,042 individuals without known heart disease referred for CAC scoring, the frequency of a CAC score of 0 was only modestly lower in smokers (38%) compared with nonsmokers (42%), yet the all-cause mortality rate was significantly higher.30 In addition, Multi-Ethnic Study of Atherosclerosis (MESA) participants who were current smokers or eligible for LCS and had a CAC score of 0 had an observed 11-year ASCVD event rate of 13.4% and 20.8%, respectively, leading to the conclusion that a CAC score of 0 may not be predictive of minimal risk in smokers and those eligible for LCS.31 Additionally, in LCS-eligible individuals, the PCE underestimated event rates and incorporation of CAC scores did not significantly improve risk estimation. Finally, data from the NLST screening trial showed that the absence of CAC on LDCT was not associated with better survival or lower CV mortality compared with individuals with low CAC scores.32

 

 

The question of whether individuals undergoing LCS with LDCT who have no detectable CAC can avoid statin therapy is an unresolved issue; no contemporary studies have looked specifically at the relationship between estimated risk, a CAC score of 0, and ASCVD outcomes in individuals participating in LCS. For these reasons, we recommend moderate-intensity statin therapy when the estimated risk is intermediate because it is unclear that either an Agatston score of 0 reclassifies intermediate-risk LCS-eligible individuals to a lower risk group.

For the few borderline risk (estimated risk, 5% to < 7.5%) LCS-eligible individuals, a CAC score of 0 might confer low short-term risk but the long-term benefit of statin therapy on reducing subsequent risk, the presence of other risk factors, and the willingness to stop smoking should all be considered. For these individuals who elect to avoid statin therapy, annual re-estimation of risk at the time of repeat LDCT is recommended. In these circumstances, referral for traditional Agatston scoring is not likely to change decision making because the sensitivity of the 2 techniques is very similar.

Agatston Score of 1-99 or CAC-DRS or Visual Score of 1

In general population studies, these scores correspond to borderline risk and an estimated 10-year event rate of just under 7.5%.20 In both the NELSON and NLST LCS trials, even low amounts of CAC regardless of the scoring method were associated with higher observed ASCVD mortality when adjusted for other baseline risk factors.32 Thus, in patients undergoing LCS with intermediate and borderline risk, a CAC score between 1 and 99 or a visual estimate of 1 indicates the presence of subclinical atherosclerosis, and moderate-intensity statin therapy is reasonable.

Agatston Score of 100-299 or CAC-DRS or Visual Score of 2

Across all ages, races, and sexes, CAC scores between 100 to 299 are associated with an event rate of about 15% over 10 years.20 In the NELSON LCS trial, the adjusted hazard ratio for ASCVD events with a nontraditional Agatston score of 101 to 400 was 6.58.33 Thus, in patients undergoing LCS with a CAC score of 100 to 299, regardless of the baseline risk estimate, the projected absolute event rate at 10 years would be about 20%. Moderate-intensity statin therapy is recommended to reduce the baseline LDL-C by 30% to 49%.

Agatston Score of > 300 orCAC-DRS or Visual Score of 3

Agatston CAC scores > 300 are consistent with a 10-year incidence of ASCVD events of > 15% regardless of age, sex, or race and ethnicity.20 In the Calcium Consortium, a CAC > 400 was correlated with an event rate of 13.6 events/1000 person-years.12 In a Walter Reed Military Medical Center study, a CAC score > 400 projected a cumulative incidence of ASCVD events of nearly 20% at 10 years.34 In smokers eligible for LCS, a CAC score > 300 projected a 10-year ASCVD event rate of 25%.29 In these patients, moderate-intensity statin therapy is recommended, although high-intensity statin therapy can be considered if there are other risk factors.

Agatston Score ≥ 1000

The 2018 consensus statement on CAC reporting categorizes all CAC scores > 300 into a single risk group because the recommended treatment options do not differ.19 However, recent data suggest this might not be the case since individuals with very high CAC scores experience high rates of events that might justify more aggressive intervention. In an analysis of individuals who participated in the CAC Consortium with a CAC score ≥ 1000, the all-cause mortality rate was 18.8 per 1000 person-years with a CV mortality rate of 8 per 1000 person-years.35 Individuals with very high levels of CAC > 1000 also have a greater number of diseased coronary arteries, higher involvement of the left main coronary artery, and significantly higher event rates compared with those with a CAC of 400 to 999.36 In an analysis of individuals from the NLST trial, nontraditionally measured Agatston score > 1000 was associated with a hazard ratio for coronary artery disease (CAD) mortality of 3.66 in men and 5.81 in women.17 These observed and projected levels of risk are like that seen in secondary prevention trials, and some experts have recommended the use of high-intensity statin therapy to reduce LDL-C to < 70 mg/dL.37

Primary Prevention in Individuals aged 76 to 80 years

LCS can continue through age 80 years, while the PCE and primary prevention guidelines are truncated at age 75 years. Because age is a major contributor to risk, many of these individuals will already be in the intermediate- to high-risk group. However, the net clinical benefit of statin therapy for primary prevention in this age group is not well established, and the few primary prevention trials in this group have not demonstrated net clinical benefit.38 As a result, current guidelines do not provide specific treatment recommendations for individuals aged > 75 years but recognize the value of shared decision making considering associated comorbidities, age-related risks of statin therapy, and the desires of the individual to avoid ASCVD-related events even if the net clinical benefit is low.

Older individuals with elevated CAC scores should be informed about the risk of ASCVD events and the potential but unproven benefit of moderate-intensity statin therapy. Older individuals with a CAC score of 0 likely have low short-term risk of ASCVD events and withholding statin therapy is not unreasonable.

 

 

CAC Scores on Annual LDCT Scans

Because LCS requires annual LDCT scans, primary care practitioners and patients need to understand the significance of changing CAC scores over time. For individuals not on statin therapy, increasing calcification is a marker of progression of subclinical atherosclerosis. Patients undergoing LCS not on statin who have progressive increases in their CAC should consider initiating statin therapy. Individuals who opted not to initiate statin therapy who subsequently develop CAC should be re-engaged in a discussion about the significance of the finding and the clinically proven benefits of statin therapy in individuals with subclinical atherosclerosis. These considerations do not apply to individuals already on statin therapy. Statins convert lipid-rich plaques to lipid-depleted plaques, resulting in increasing calcification. As a result, CAC scores do not decrease and may increase with statin therapy.39 Individuals participating in annual LCS should be informed of this possibility. Also, in these individuals, referral to specialty care as a treatment failure is not supported by the literature.

Furthermore, serial CAC scoring to titrate the intensity of statin therapy is not currently recommended. The goal with moderate-intensity statin therapy is a 30% to 49% reduction from baseline LDL-C. If this milestone is not achieved, the statin dose can be escalated. For high-intensity statin therapy, the goal is a > 50% reduction. If this milestone is not achieved, then additional lipid-lowering agents, such as ezetimibe, can be added.

Further ASCVD Testing

LCS with LDCT is associated with improved health outcomes, and LDCT is the preferred imaging modality. The ability of LDCT to detect and quantify CAC is sufficient for clinical decision making. Therefore, obtaining a traditional CAC score increases radiation exposure without additional clinical benefits.

Furthermore, although referral for additional testing in those with nonzero CAC scores is common, current evidence does not support this practice in asymptomatic individuals. Indeed, the risks of LCS include overdiagnosis, excessive testing, and overtreatment secondary to the discovery of other findings, such as benign pulmonary nodules and CAC. With respect to CAD, randomized controlled trials do not support a strategy of coronary angiography and intervention in asymptomatic individuals, even with moderate-to-severe ischemia on functional testing.40 As a result, routine stress tests to diagnose CAD or to confirm the results of CAC scores in asymptomatic individuals are not recommended. The only potential exception would be in select cases where the CAC score is > 1000 and when calcium is predominately located in the left main coronary artery.

Conclusions

LCS provides smokers at risk for lung cancer with the best probability to survive that diagnosis, and coincidentally LCS may also provide the best opportunity to prevent ASCVD events and mortality. Before initiating LCS, clinicians should initiate a shared decision making conversation about the benefits and risks of LDCT scans. In addition to relevant education about smoking, during shared decision making, the initial ASCVD risk estimate should be done using the PCE and when appropriate the benefits of statin therapy discussed. Individuals also should be informed of the potential for identifying CAC and counseled on its significance and how it might influence the decision to recommend statin therapy.

In patients undergoing LCS with an estimated risk of ≥ 7.5% to < 20%, moderate-intensity statin therapy is indicated. In this setting, a CAC score > 0 indicates subclinical atherosclerosis and should be used to help direct patients toward initiating statin therapy. Unfortunately, in patients undergoing LCS a CAC score of 0 might not provide protection against ASCVD, and until there is more information to the contrary, these individuals should at least participate in shared decision making about the long-term benefits of statin therapy in reducing ASCVD risk. Because LDCT scanning is done annually, there are opportunities to review the importance of prevention and to adjust therapy as needed to achieve the greatest reduction in ASCVD. Reported elevated CAC scores on LDCT provide an opportunity to re-engage the patient in the discussion about the benefits of statin therapy if they are not already on a statin, or consideration for high-intensity statin if the CAC score is > 1000 or reduction in baseline LDL-C is < 30% on the current statin dose.

 

Lung cancer is the most common cause of cancer mortality, and cigarette smoking is the most significant risk factor. Several randomized clinical trials have shown that lung cancer screening (LCS) with nonelectrocardiogram (ECG)-gated low-dose computed tomography (LDCT) reduces both lung cancer and all-cause mortality.1,2 Hence, the US Preventive Screening Task Force (USPSTF) recommends annual screening with LDCT in adults aged 50 to 80 years who have a 20-pack-year smoking history and currently smoke or have quit within the past 15 years.3

Smoking is also an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), and LCS clinical trials acknowledge that mortality from ASCVD events exceeds that of lung cancer.4,5 In an analysis of asymptomatic individuals from the Framingham Heart Offspring study who were eligible for LCS, the ASCVD event rate during a median (IQR) follow-up of 11.4 (9.7-12.0) years was 12.6%.6 However, despite the high rate of ASCVD events in this population, primary prevention strategies are consistently underused. In a study of 5495 individuals who underwent LCS with LDCT, only 40% of those eligible for statins had one prescribed, underscoring the missed opportunity for preventing ASCVD events during LCS.7 Yet the interactions for shared decision making and the availability of coronary artery calcification (CAC) scores from the LDCT provide an ideal window for intervening and preventing ASCVD events during LCS.

CAC is a hallmark of atherosclerotic plaque development and is proportional to plaque burden and ASCVD risk.8 Because of the relationship between CAC, subclinical atherosclerosis, and ASCVD risk, there is an opportunity to use CAC detected by LDCT to predict ASCVD risk and guide recommendations for statin treatment in individuals enrolled in LCS. Traditionally, CAC has been visualized by ECG-gated noncontrast CT scans with imaging protocols specifically designed to visualize the coronary arteries, minimize motion artifacts, and reduce signal noise. These scans are specifically done for primary prevention risk assessment and report an Agatston score, a summed measure based on calcified plaque area and maximal density.9 Results are reported as an overall CAC score and an age-, sex-, and race-adjusted percentile of CAC. Currently, a CAC score ≥ 100 or above the 75th percentile for age, sex, and race is considered abnormal.

High-quality evidence supports CAC scores as a strong predictor of ASCVD risk independent of age, sex, race, and other traditional risk factors.10-12 In asymptomatic individuals, a CAC score of 0 is a strong, negative risk factor associated with very low annualized mortality rates and cardiovascular (CV) events, so intermediate-risk individuals can be reclassified to a lower risk group avoiding or delaying statin therapy.13 As a result, current primary prevention guidelines allow for CAC scoring in asymptomatic, intermediate-risk adults where the clinical benefits of statin therapy are uncertain, knowing the CAC score will aid in the clinical decision to delay or initiate statin therapy.

Unlike traditional ECG-gated CAC scoring, LDCT imaging protocols are non–ECG-gated and performed at variable energy and slice thickness to optimize the detection of lung nodules. Early studies suggested that CAC detected by LDCT could be used in lieu of traditional CAC scoring to personalize risk.14,15 Recently, multiple studies have validated the accuracy and reproducibility of LDCT to detect and quantify CAC. In both the NELSON and the National Lung Screening Trial (NLST) LCS trials, higher visual and quantitative measures of CAC were independently and incrementally associated with ASCVD risk.16,17 A subsequent review and meta-analysis of 6 LCS trials confirmed CAC detected by LDCT to be an independent predictor of ASCVD events regardless of the method used to measure CAC.18

table.png

There is now consensus that either an Agatston score or a visual estimate of CAC be reported on all noncontrast, noncardiac chest CT scans irrespective of the indication or technique, including LDCT scans for LCS using a uniform reporting system known as the Coronary Artery Calcium Data and Reporting System (CAC-DRS).19 The CAC-DRS simplifies reporting and adds modifiers indicating if the reported score is visual (V) or Agatston (A) and number of vessels involved. For example, CAC-DRS A0 or CAC-DRS V0 would indicate an Agatston score of 0 or a visual score of 0. CAC-DRS A1/N2 would indicate a total Agatston score of 1-99 in 2 coronary arteries. The currently agreed-on CAC-DRS risk groups are listed in the Table, along with their corresponding visual score or Agatston score and anticipated 10-year event rate, irrespective of other risk factors.20

As LCS efforts increase, primary care practitioners will receive LDCT reports that now incorporate an estimation of CAC (visual or quantitative). Thus, it will be increasingly important to know how to interpret and use these scores to guide clinical decisions regarding the initiation of statin therapy, referral for additional testing, and when to seek specialty cardiology care. For instance, does the absence of CAC (CAC = 0) on LDCT predict a low enough risk for statin therapy to be delayed or withdrawn? Does increasing CAC scores on follow-up LDCT in individuals on statin therapy represent treatment failure? When should CAC scores trigger additional testing, such as a stress test or referral to cardiology specialty care?

 

 

Primary Prevention in LCS

The initial approach to primary prevention in LCS is no different from that recommended by the 2018 multisociety guidelines on the management of blood cholesterol, the 2019 American College of Cardiology/American Heart Association (ACC/AHA) guideline on primary prevention, or the 2022 USPTSF recommendations on statin use for primary prevention of CV disease in adults.21-23 For a baseline low-density lipoprotein cholesterol (LDL-C) ≥ 190 mg/dL, high-intensity statin therapy is recommended without further risk stratification. Individuals with diabetes also are at higher-than-average risk, and moderate-intensity statin therapy is recommended.

For individuals not in either group, a validated ASCVD risk assessment tool is recommended to estimate baseline risk. The most validated tool for estimating risk in the US population is the 2013 ACC/AHA Pooled Cohort Equation (PCE) which provides an estimate of the 10-year risk for fatal and myocardial infarction and fatal and nonfatal stroke.24 The PCE risk calculator uses age, presence of diabetes, sex, smoking history, total cholesterol, high-density lipoprotein cholesterol, systolic blood pressure, and treatment for hypertension to place individuals into 1 of 4 risk groups: low (< 5%), borderline (5% to < 7.5%), intermediate (≥ 7.5% to < 20%), and high (≥ 20%). Clinicians should be aware that the PCE only considers current smoking history and not prior smoking history or cumulative pack-year history. This differs from eligibility for LCS where recent smoking plays a larger role. All these risk factors are important to consider when evaluating risk and discussing risk-reducing strategies like statin therapy.

The 2018 multisociety guidelines and the 2019 primary prevention guidelines set the threshold for considering initiation of statin therapy at intermediate risk ≥ 7.5%.21,22 The 2020 US Department of Veterans Affairs/Department of Defense guidelines set the threshold for considering statin therapy at an estimated 10-year event rate of 12%, whereas the 2022 UPSTF recommendations set the threshold at 10% with additional risk factors as the threshold for statin therapy.23,25 The reasons for these differences are beyond the scope of this review, but all these guidelines use the PCE to estimate baseline risk as the starting point for clinical decision making.

The PCE was originally derived and validated in population studies dating to the 1960s when the importance of diet, exercise, and smoking cessation in reducing ASCVD events was not well appreciated. The application of the PCE in more contemporary populations shows that it overestimates risk, especially in older individuals and women.26,27 Overestimation of risk has the potential to result in the initiation of statin therapy in individuals in whom the actual clinical benefit would otherwise be small.

figure.png

To address this issue, current guidelines allow the use of CAC scoring to refine risk in individuals who are classified as intermediate risk and who otherwise desire to avoid lifelong statin therapy. Using current recommendations, we make suggestions on how to use CAC scores from LDCT to aid in clinical decision making for individuals in LCS (Figure).

No Coronary Artery Calcification

Between 25% and 30% of LDCT done for LCS will show no CAC.14,16 In general population studies, a CAC score of 0 is a strong negative predictor when there are no other risk factors.13,28 In contrast, the negative predictive ability of a CAC score of 0 in individuals with a smoking history who are eligible for LCS is unproven. In multivariate modeling, a CAC score of 0 did not reduce the significant hazard of all-cause mortality in patients with diabetes or smokers.29 In an analysis of 44,042 individuals without known heart disease referred for CAC scoring, the frequency of a CAC score of 0 was only modestly lower in smokers (38%) compared with nonsmokers (42%), yet the all-cause mortality rate was significantly higher.30 In addition, Multi-Ethnic Study of Atherosclerosis (MESA) participants who were current smokers or eligible for LCS and had a CAC score of 0 had an observed 11-year ASCVD event rate of 13.4% and 20.8%, respectively, leading to the conclusion that a CAC score of 0 may not be predictive of minimal risk in smokers and those eligible for LCS.31 Additionally, in LCS-eligible individuals, the PCE underestimated event rates and incorporation of CAC scores did not significantly improve risk estimation. Finally, data from the NLST screening trial showed that the absence of CAC on LDCT was not associated with better survival or lower CV mortality compared with individuals with low CAC scores.32

 

 

The question of whether individuals undergoing LCS with LDCT who have no detectable CAC can avoid statin therapy is an unresolved issue; no contemporary studies have looked specifically at the relationship between estimated risk, a CAC score of 0, and ASCVD outcomes in individuals participating in LCS. For these reasons, we recommend moderate-intensity statin therapy when the estimated risk is intermediate because it is unclear that either an Agatston score of 0 reclassifies intermediate-risk LCS-eligible individuals to a lower risk group.

For the few borderline risk (estimated risk, 5% to < 7.5%) LCS-eligible individuals, a CAC score of 0 might confer low short-term risk but the long-term benefit of statin therapy on reducing subsequent risk, the presence of other risk factors, and the willingness to stop smoking should all be considered. For these individuals who elect to avoid statin therapy, annual re-estimation of risk at the time of repeat LDCT is recommended. In these circumstances, referral for traditional Agatston scoring is not likely to change decision making because the sensitivity of the 2 techniques is very similar.

Agatston Score of 1-99 or CAC-DRS or Visual Score of 1

In general population studies, these scores correspond to borderline risk and an estimated 10-year event rate of just under 7.5%.20 In both the NELSON and NLST LCS trials, even low amounts of CAC regardless of the scoring method were associated with higher observed ASCVD mortality when adjusted for other baseline risk factors.32 Thus, in patients undergoing LCS with intermediate and borderline risk, a CAC score between 1 and 99 or a visual estimate of 1 indicates the presence of subclinical atherosclerosis, and moderate-intensity statin therapy is reasonable.

Agatston Score of 100-299 or CAC-DRS or Visual Score of 2

Across all ages, races, and sexes, CAC scores between 100 to 299 are associated with an event rate of about 15% over 10 years.20 In the NELSON LCS trial, the adjusted hazard ratio for ASCVD events with a nontraditional Agatston score of 101 to 400 was 6.58.33 Thus, in patients undergoing LCS with a CAC score of 100 to 299, regardless of the baseline risk estimate, the projected absolute event rate at 10 years would be about 20%. Moderate-intensity statin therapy is recommended to reduce the baseline LDL-C by 30% to 49%.

Agatston Score of > 300 orCAC-DRS or Visual Score of 3

Agatston CAC scores > 300 are consistent with a 10-year incidence of ASCVD events of > 15% regardless of age, sex, or race and ethnicity.20 In the Calcium Consortium, a CAC > 400 was correlated with an event rate of 13.6 events/1000 person-years.12 In a Walter Reed Military Medical Center study, a CAC score > 400 projected a cumulative incidence of ASCVD events of nearly 20% at 10 years.34 In smokers eligible for LCS, a CAC score > 300 projected a 10-year ASCVD event rate of 25%.29 In these patients, moderate-intensity statin therapy is recommended, although high-intensity statin therapy can be considered if there are other risk factors.

Agatston Score ≥ 1000

The 2018 consensus statement on CAC reporting categorizes all CAC scores > 300 into a single risk group because the recommended treatment options do not differ.19 However, recent data suggest this might not be the case since individuals with very high CAC scores experience high rates of events that might justify more aggressive intervention. In an analysis of individuals who participated in the CAC Consortium with a CAC score ≥ 1000, the all-cause mortality rate was 18.8 per 1000 person-years with a CV mortality rate of 8 per 1000 person-years.35 Individuals with very high levels of CAC > 1000 also have a greater number of diseased coronary arteries, higher involvement of the left main coronary artery, and significantly higher event rates compared with those with a CAC of 400 to 999.36 In an analysis of individuals from the NLST trial, nontraditionally measured Agatston score > 1000 was associated with a hazard ratio for coronary artery disease (CAD) mortality of 3.66 in men and 5.81 in women.17 These observed and projected levels of risk are like that seen in secondary prevention trials, and some experts have recommended the use of high-intensity statin therapy to reduce LDL-C to < 70 mg/dL.37

Primary Prevention in Individuals aged 76 to 80 years

LCS can continue through age 80 years, while the PCE and primary prevention guidelines are truncated at age 75 years. Because age is a major contributor to risk, many of these individuals will already be in the intermediate- to high-risk group. However, the net clinical benefit of statin therapy for primary prevention in this age group is not well established, and the few primary prevention trials in this group have not demonstrated net clinical benefit.38 As a result, current guidelines do not provide specific treatment recommendations for individuals aged > 75 years but recognize the value of shared decision making considering associated comorbidities, age-related risks of statin therapy, and the desires of the individual to avoid ASCVD-related events even if the net clinical benefit is low.

Older individuals with elevated CAC scores should be informed about the risk of ASCVD events and the potential but unproven benefit of moderate-intensity statin therapy. Older individuals with a CAC score of 0 likely have low short-term risk of ASCVD events and withholding statin therapy is not unreasonable.

 

 

CAC Scores on Annual LDCT Scans

Because LCS requires annual LDCT scans, primary care practitioners and patients need to understand the significance of changing CAC scores over time. For individuals not on statin therapy, increasing calcification is a marker of progression of subclinical atherosclerosis. Patients undergoing LCS not on statin who have progressive increases in their CAC should consider initiating statin therapy. Individuals who opted not to initiate statin therapy who subsequently develop CAC should be re-engaged in a discussion about the significance of the finding and the clinically proven benefits of statin therapy in individuals with subclinical atherosclerosis. These considerations do not apply to individuals already on statin therapy. Statins convert lipid-rich plaques to lipid-depleted plaques, resulting in increasing calcification. As a result, CAC scores do not decrease and may increase with statin therapy.39 Individuals participating in annual LCS should be informed of this possibility. Also, in these individuals, referral to specialty care as a treatment failure is not supported by the literature.

Furthermore, serial CAC scoring to titrate the intensity of statin therapy is not currently recommended. The goal with moderate-intensity statin therapy is a 30% to 49% reduction from baseline LDL-C. If this milestone is not achieved, the statin dose can be escalated. For high-intensity statin therapy, the goal is a > 50% reduction. If this milestone is not achieved, then additional lipid-lowering agents, such as ezetimibe, can be added.

Further ASCVD Testing

LCS with LDCT is associated with improved health outcomes, and LDCT is the preferred imaging modality. The ability of LDCT to detect and quantify CAC is sufficient for clinical decision making. Therefore, obtaining a traditional CAC score increases radiation exposure without additional clinical benefits.

Furthermore, although referral for additional testing in those with nonzero CAC scores is common, current evidence does not support this practice in asymptomatic individuals. Indeed, the risks of LCS include overdiagnosis, excessive testing, and overtreatment secondary to the discovery of other findings, such as benign pulmonary nodules and CAC. With respect to CAD, randomized controlled trials do not support a strategy of coronary angiography and intervention in asymptomatic individuals, even with moderate-to-severe ischemia on functional testing.40 As a result, routine stress tests to diagnose CAD or to confirm the results of CAC scores in asymptomatic individuals are not recommended. The only potential exception would be in select cases where the CAC score is > 1000 and when calcium is predominately located in the left main coronary artery.

Conclusions

LCS provides smokers at risk for lung cancer with the best probability to survive that diagnosis, and coincidentally LCS may also provide the best opportunity to prevent ASCVD events and mortality. Before initiating LCS, clinicians should initiate a shared decision making conversation about the benefits and risks of LDCT scans. In addition to relevant education about smoking, during shared decision making, the initial ASCVD risk estimate should be done using the PCE and when appropriate the benefits of statin therapy discussed. Individuals also should be informed of the potential for identifying CAC and counseled on its significance and how it might influence the decision to recommend statin therapy.

In patients undergoing LCS with an estimated risk of ≥ 7.5% to < 20%, moderate-intensity statin therapy is indicated. In this setting, a CAC score > 0 indicates subclinical atherosclerosis and should be used to help direct patients toward initiating statin therapy. Unfortunately, in patients undergoing LCS a CAC score of 0 might not provide protection against ASCVD, and until there is more information to the contrary, these individuals should at least participate in shared decision making about the long-term benefits of statin therapy in reducing ASCVD risk. Because LDCT scanning is done annually, there are opportunities to review the importance of prevention and to adjust therapy as needed to achieve the greatest reduction in ASCVD. Reported elevated CAC scores on LDCT provide an opportunity to re-engage the patient in the discussion about the benefits of statin therapy if they are not already on a statin, or consideration for high-intensity statin if the CAC score is > 1000 or reduction in baseline LDL-C is < 30% on the current statin dose.

References

1. de Koning HJ, van der Aalst CM, Oudkerk M. Lung-cancer screening and the NELSON Trial. Reply. N Engl J Med. 2020;382(22):2165-2166. doi:10.1056/NEJMc2004224

2. Aberle T, Adams DR, Berg AM, et al; National Lung Screening Trial Research Team. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):396-409. doi:10.1056/NEJMoa1102873

3. Krist AH, Davidson KW, Mangione CM, et al; US Preventive Services Task Force. Screening for lung cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;25(10):962-970. doi:10.1001/jama.2021.1117

4. Jha P, Ramasundarahettige C, Landsman V. 21st-century hazards of smoking and benefits of cessation in the United States. N Engl J Med. 2013;368(4):341-350. doi:10.1056/NEJMsa1211128

5. Khan SS, Ning H, Sinha A, et al. Cigarette smoking and competing risks for fatal and nonfatal cardiovascular disease subtypes across the life course. J Am Heart Assoc. 2021;10(23):e021751. doi:10.1161/JAHA.121.021751

6. Lu MT, Onuma OK, Massaro JM, et al. Lung cancer screening eligibility in the community: cardiovascular risk factors, coronary artery calcification, and cardiovascular events. Circulation. 2016;134(12):897-899. doi:10.1161/CIRCULATIONAHA.116.023957

7. Tailor TD, Chiles C, Yeboah J, et al. Cardiovascular risk in the lung cancer screening population: a multicenter study evaluating the association between coronary artery calcification and preventive statin prescription. J Am Coll Radiol. 2021;18(9):1258-1266. doi:10.1016/j.jacr.2021.01.015

8. Mori H, Torii S, Kutyna M, et al. Coronary artery calcification and its progression: what does it really mean? JACC Cardiovasc Imaging. 2018;11(1):127-142. doi:10.1016/j.jcmg.2017.10.012

10. Nasir K, Bittencourt MS, Blaha MJ, et al. Implications of coronary artery calcium testing among statin candidates according to American College of Cardiology/American Heart Association cholesterol management guidelines: MESA (Multi-Ethnic Study of Atherosclerosis). J Am Coll Cardiol. 2015;66(15): 1657-1668. doi:10.1016/j.jacc.2015.07.066

11. Detrano R, Guerci AD, Carr JJ, et al. Coronary calcium as a predictor of coronary events in four racial or ethnic groups. N Engl J Med. 2008;358(13):1336-1345. doi:10.1056/NEJMoa072100

12. Grandhi GR, Mirbolouk M, Dardari ZA. Interplay of coronary artery calcium and risk factors for predicting CVD/CHD Mortality: the CAC Consortium. JACC Cardiovasc Imaging. 2020;13(5):1175-1186. doi:10.1016/j.jcmg.2019.08.024

13. Blaha M, Budoff MJ, Shaw J. Absence of coronary artery calcification and all-cause mortality. JACC Cardiovasc Imaging. 2009;2(6):692-700. doi:10.1016/j.jcmg.2009.03.009

14. Shemesh J, Henschke CI, Farooqi A, et al. Frequency of coronary artery calcification on low-dose computed tomography screening for lung cancer. Clin Imaging. 2006;30(3):181-185. doi:10.1016/j.clinimag.2005.11.002

15. Shemesh J, Henschke C, Shaham D, et al. Ordinal scoring of coronary artery calcifications on low-dose CT scans of the chest is predictive of death from cardiovascular disease. Radiology. 2010;257:541-548. doi:10.1148/radiol.10100383

16. Jacobs PC, Gondrie MJ, van der Graaf Y, et al. Coronary artery calcium can predict all-cause mortality and cardiovascular events on low-dose CT screening for lung cancer. AJR Am J Roentgenol. 2012;198(3):505-511. doi:10.2214/AJR.10.5577

17. Lessmann N, de Jong PA, Celeng C, et al. Sex differences in coronary artery and thoracic aorta calcification and their association with cardiovascular mortality in heavy smokers. JACC Cardiovasc Imaging. 2019;12(9):1808-1817. doi:10.1016/j.jcmg.2018.10.026

18. Gendarme S, Goussault H, Assie JB, et al. Impact on all-cause and cardiovascular mortality rates of coronary artery calcifications detected during organized, low-dose, computed-tomography screening for lung cancer: systematic literature review and meta-analysis. Cancers (Basel). 2021;13(7):1553. doi:10.3390/cancers13071553

19. Hecht HS, Blaha MJ, Kazerooni EA, et al. CAC-DRS: coronary artery calcium data and reporting system. An expert consensus document of the Society of Cardiovascular Computed Tomography (SCCT). J Cardiovasc Comput Tomogr. 2018;12(3):185-191. doi:10.1016/j.jcct.2018.03.008

20. Budoff MJ, Young R, Burke G, et al. Ten-year association of coronary artery calcium with atherosclerotic cardiovascular disease (ASCVD) events: the multi-ethnic study of atherosclerosis (MESA). Eur Heart J. 2018;39(25):2401-2408. doi:10.1093/eurheartj/ehy217

21. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139(25):e1046-e1081. doi:10.1161/CIR.0000000000000624

22. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678

23. Mangione CM, Barry MJ, Nicholson WK, et al; US Preventive Services Task Force. Statin use for the primary prevention of cardiovascular disease in adults: US Preventive Services Task Force recommendation statement. JAMA. 2022;328(8):746-753. doi:10.1001/jama.2022.13044

24. Stone NJ, Robinson JG, Lichtenstein AH, et al; American College of Cardiology/American Heart Association Task Force on Practice. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63(25 pt B):2889-2934. doi:10.1016/j.jacc.2013.11.002

<--pagebreak-->

25. US Department of Veterans Affairs, Department of Defense. VA/DoD clinical practice guideline. Updated August 25, 2021. Accessed November 3, 2023. https://www.healthquality.va.gov/guidelines/cd/lipids

26. DeFilippis AP, Young, R, Carrubba CJ, et al. An analysis of calibration and discrimination among multiple cardiovascular risk scores in a modern multiethnic cohort. Ann Intern Med. 2015;162(4):266-275. doi:10.7326/M14-1281

27. Rana JS, Tabada GH, Solomon, MD, et al. Accuracy of the atherosclerotic cardiovascular risk equation in a large contemporary, multiethnic population. J Am Coll Cardiol. 2016;67(18):2118-2130. doi:10.1016/j.jacc.2016.02.055

28. Sarwar A, Shaw LJ, Shapiro MD, et al. Diagnostic and prognostic value of absence of coronary artery calcification. JACC Cardiovasc Imaging. 2009;2(6):675-688. doi:10.1016/j.jcmg.2008.12.031

29. McEvoy JW, Blaha MJ, Rivera JJ, et al. Mortality rates in smokers and nonsmokers in the presence or absence of coronary artery calcification. JACC Cardiovasc Imaging. 2012;5(10):1037-1045. doi:10.1016/j.jcmg.2012.02.017

30. Leigh A, McEvoy JW, Garg P, et al. Coronary artery calcium scores and atherosclerotic cardiovascular disease risk stratification in smokers. JACC Cardiovasc Imaging. 2019;12(5):852-861. doi:10.1016/j.jcmg.2017.12.017

31. Garg PK, Jorgensen NW, McClelland RL, et al. Use of coronary artery calcium testing to improve coronary heart disease risk assessment in lung cancer screening population: The Multi-Ethnic Study of Atherosclerosis (MESA). J Cardiovasc Comput Tomagr. 2018;12(6):439-400.

32. Chiles C, Duan F, Gladish GW, et al. Association of coronary artery calcification and mortality in the national lung screening trial: a comparison of three scoring methods. Radiology. 2015;276(1):82-90. doi:10.1148/radiol.15142062

33. Takx RA, Isgum I, Willemink MJ, et al. Quantification of coronary artery calcium in nongated CT to predict cardiovascular events in male lung cancer screening participants: results of the NELSON study. J Cardiovasc Comput Tomogr. 2015;9(1):50-57. doi:10.1016/j.jcct.2014.11.006

34. Mitchell JD, Paisley R, Moon P, et al. Coronary artery calcium and long-term risk of death, myocardial infarction, and stroke: The Walter Reed Cohort Study. JACC Cardiovasc Imaging. 2018;11(12):1799-1806. doi:10.1016/j.jcmg.2017.09.003

35. Peng AW, Mirbolouk M, Orimoloye OA, et al. Long-term all-cause and cause-specific mortality in asymptomatic patients with CAC >/=1,000: results from the CAC Consortium. JACC Cardiovasc Imaging. 2019;13(1, pt 1):83-93. doi:10.1016/j.jcmg.2019.02.005

36. Peng AW, Dardari ZA. Blumenthal RS, et al. Very high coronary artery calcium (>/=1000) and association with cardiovascular disease events, non-cardiovascular disease outcomes, and mortality: results from MESA. Circulation. 2021;143(16):1571-1583. doi:10.1161/CIRCULATIONAHA.120.050545

37. Orringer CE, Blaha MJ, Blankstein R, et al. The National Lipid Association scientific statement on coronary artery calcium scoring to guide preventive strategies for ASCVD risk reduction. J Clin Lipidol. 2021;15(1):33-60. doi:10.1016/j.jacl.2020.12.005

38. Sheperd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease. (PROSPER): a randomized controlled trial. Lancet. 2002;360:1623-1630. doi:10.1016/s0140-6736(02)11600-x

39. Puri R, Nicholls SJ, Shao M, et al. Impact of statins on serial coronary calcification during atheroma progression and regression. J Am Coll Cardiol. 2015;65(13):1273-1282. doi:10.1016/j.jacc.2015.01.036

40. Maron D.J, Hochman J S, Reynolds HR, et al; ISCHEMIA Research Group. Initial invasive or conservative strategy for stable coronary disease. N Engl J Med. 2020;382(15):1395-1407. doi:10.1056/NEJMoa1915922

References

1. de Koning HJ, van der Aalst CM, Oudkerk M. Lung-cancer screening and the NELSON Trial. Reply. N Engl J Med. 2020;382(22):2165-2166. doi:10.1056/NEJMc2004224

2. Aberle T, Adams DR, Berg AM, et al; National Lung Screening Trial Research Team. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):396-409. doi:10.1056/NEJMoa1102873

3. Krist AH, Davidson KW, Mangione CM, et al; US Preventive Services Task Force. Screening for lung cancer: US Preventive Services Task Force recommendation statement. JAMA. 2021;25(10):962-970. doi:10.1001/jama.2021.1117

4. Jha P, Ramasundarahettige C, Landsman V. 21st-century hazards of smoking and benefits of cessation in the United States. N Engl J Med. 2013;368(4):341-350. doi:10.1056/NEJMsa1211128

5. Khan SS, Ning H, Sinha A, et al. Cigarette smoking and competing risks for fatal and nonfatal cardiovascular disease subtypes across the life course. J Am Heart Assoc. 2021;10(23):e021751. doi:10.1161/JAHA.121.021751

6. Lu MT, Onuma OK, Massaro JM, et al. Lung cancer screening eligibility in the community: cardiovascular risk factors, coronary artery calcification, and cardiovascular events. Circulation. 2016;134(12):897-899. doi:10.1161/CIRCULATIONAHA.116.023957

7. Tailor TD, Chiles C, Yeboah J, et al. Cardiovascular risk in the lung cancer screening population: a multicenter study evaluating the association between coronary artery calcification and preventive statin prescription. J Am Coll Radiol. 2021;18(9):1258-1266. doi:10.1016/j.jacr.2021.01.015

8. Mori H, Torii S, Kutyna M, et al. Coronary artery calcification and its progression: what does it really mean? JACC Cardiovasc Imaging. 2018;11(1):127-142. doi:10.1016/j.jcmg.2017.10.012

10. Nasir K, Bittencourt MS, Blaha MJ, et al. Implications of coronary artery calcium testing among statin candidates according to American College of Cardiology/American Heart Association cholesterol management guidelines: MESA (Multi-Ethnic Study of Atherosclerosis). J Am Coll Cardiol. 2015;66(15): 1657-1668. doi:10.1016/j.jacc.2015.07.066

11. Detrano R, Guerci AD, Carr JJ, et al. Coronary calcium as a predictor of coronary events in four racial or ethnic groups. N Engl J Med. 2008;358(13):1336-1345. doi:10.1056/NEJMoa072100

12. Grandhi GR, Mirbolouk M, Dardari ZA. Interplay of coronary artery calcium and risk factors for predicting CVD/CHD Mortality: the CAC Consortium. JACC Cardiovasc Imaging. 2020;13(5):1175-1186. doi:10.1016/j.jcmg.2019.08.024

13. Blaha M, Budoff MJ, Shaw J. Absence of coronary artery calcification and all-cause mortality. JACC Cardiovasc Imaging. 2009;2(6):692-700. doi:10.1016/j.jcmg.2009.03.009

14. Shemesh J, Henschke CI, Farooqi A, et al. Frequency of coronary artery calcification on low-dose computed tomography screening for lung cancer. Clin Imaging. 2006;30(3):181-185. doi:10.1016/j.clinimag.2005.11.002

15. Shemesh J, Henschke C, Shaham D, et al. Ordinal scoring of coronary artery calcifications on low-dose CT scans of the chest is predictive of death from cardiovascular disease. Radiology. 2010;257:541-548. doi:10.1148/radiol.10100383

16. Jacobs PC, Gondrie MJ, van der Graaf Y, et al. Coronary artery calcium can predict all-cause mortality and cardiovascular events on low-dose CT screening for lung cancer. AJR Am J Roentgenol. 2012;198(3):505-511. doi:10.2214/AJR.10.5577

17. Lessmann N, de Jong PA, Celeng C, et al. Sex differences in coronary artery and thoracic aorta calcification and their association with cardiovascular mortality in heavy smokers. JACC Cardiovasc Imaging. 2019;12(9):1808-1817. doi:10.1016/j.jcmg.2018.10.026

18. Gendarme S, Goussault H, Assie JB, et al. Impact on all-cause and cardiovascular mortality rates of coronary artery calcifications detected during organized, low-dose, computed-tomography screening for lung cancer: systematic literature review and meta-analysis. Cancers (Basel). 2021;13(7):1553. doi:10.3390/cancers13071553

19. Hecht HS, Blaha MJ, Kazerooni EA, et al. CAC-DRS: coronary artery calcium data and reporting system. An expert consensus document of the Society of Cardiovascular Computed Tomography (SCCT). J Cardiovasc Comput Tomogr. 2018;12(3):185-191. doi:10.1016/j.jcct.2018.03.008

20. Budoff MJ, Young R, Burke G, et al. Ten-year association of coronary artery calcium with atherosclerotic cardiovascular disease (ASCVD) events: the multi-ethnic study of atherosclerosis (MESA). Eur Heart J. 2018;39(25):2401-2408. doi:10.1093/eurheartj/ehy217

21. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;139(25):e1046-e1081. doi:10.1161/CIR.0000000000000624

22. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation. 2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678

23. Mangione CM, Barry MJ, Nicholson WK, et al; US Preventive Services Task Force. Statin use for the primary prevention of cardiovascular disease in adults: US Preventive Services Task Force recommendation statement. JAMA. 2022;328(8):746-753. doi:10.1001/jama.2022.13044

24. Stone NJ, Robinson JG, Lichtenstein AH, et al; American College of Cardiology/American Heart Association Task Force on Practice. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63(25 pt B):2889-2934. doi:10.1016/j.jacc.2013.11.002

<--pagebreak-->

25. US Department of Veterans Affairs, Department of Defense. VA/DoD clinical practice guideline. Updated August 25, 2021. Accessed November 3, 2023. https://www.healthquality.va.gov/guidelines/cd/lipids

26. DeFilippis AP, Young, R, Carrubba CJ, et al. An analysis of calibration and discrimination among multiple cardiovascular risk scores in a modern multiethnic cohort. Ann Intern Med. 2015;162(4):266-275. doi:10.7326/M14-1281

27. Rana JS, Tabada GH, Solomon, MD, et al. Accuracy of the atherosclerotic cardiovascular risk equation in a large contemporary, multiethnic population. J Am Coll Cardiol. 2016;67(18):2118-2130. doi:10.1016/j.jacc.2016.02.055

28. Sarwar A, Shaw LJ, Shapiro MD, et al. Diagnostic and prognostic value of absence of coronary artery calcification. JACC Cardiovasc Imaging. 2009;2(6):675-688. doi:10.1016/j.jcmg.2008.12.031

29. McEvoy JW, Blaha MJ, Rivera JJ, et al. Mortality rates in smokers and nonsmokers in the presence or absence of coronary artery calcification. JACC Cardiovasc Imaging. 2012;5(10):1037-1045. doi:10.1016/j.jcmg.2012.02.017

30. Leigh A, McEvoy JW, Garg P, et al. Coronary artery calcium scores and atherosclerotic cardiovascular disease risk stratification in smokers. JACC Cardiovasc Imaging. 2019;12(5):852-861. doi:10.1016/j.jcmg.2017.12.017

31. Garg PK, Jorgensen NW, McClelland RL, et al. Use of coronary artery calcium testing to improve coronary heart disease risk assessment in lung cancer screening population: The Multi-Ethnic Study of Atherosclerosis (MESA). J Cardiovasc Comput Tomagr. 2018;12(6):439-400.

32. Chiles C, Duan F, Gladish GW, et al. Association of coronary artery calcification and mortality in the national lung screening trial: a comparison of three scoring methods. Radiology. 2015;276(1):82-90. doi:10.1148/radiol.15142062

33. Takx RA, Isgum I, Willemink MJ, et al. Quantification of coronary artery calcium in nongated CT to predict cardiovascular events in male lung cancer screening participants: results of the NELSON study. J Cardiovasc Comput Tomogr. 2015;9(1):50-57. doi:10.1016/j.jcct.2014.11.006

34. Mitchell JD, Paisley R, Moon P, et al. Coronary artery calcium and long-term risk of death, myocardial infarction, and stroke: The Walter Reed Cohort Study. JACC Cardiovasc Imaging. 2018;11(12):1799-1806. doi:10.1016/j.jcmg.2017.09.003

35. Peng AW, Mirbolouk M, Orimoloye OA, et al. Long-term all-cause and cause-specific mortality in asymptomatic patients with CAC >/=1,000: results from the CAC Consortium. JACC Cardiovasc Imaging. 2019;13(1, pt 1):83-93. doi:10.1016/j.jcmg.2019.02.005

36. Peng AW, Dardari ZA. Blumenthal RS, et al. Very high coronary artery calcium (>/=1000) and association with cardiovascular disease events, non-cardiovascular disease outcomes, and mortality: results from MESA. Circulation. 2021;143(16):1571-1583. doi:10.1161/CIRCULATIONAHA.120.050545

37. Orringer CE, Blaha MJ, Blankstein R, et al. The National Lipid Association scientific statement on coronary artery calcium scoring to guide preventive strategies for ASCVD risk reduction. J Clin Lipidol. 2021;15(1):33-60. doi:10.1016/j.jacl.2020.12.005

38. Sheperd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease. (PROSPER): a randomized controlled trial. Lancet. 2002;360:1623-1630. doi:10.1016/s0140-6736(02)11600-x

39. Puri R, Nicholls SJ, Shao M, et al. Impact of statins on serial coronary calcification during atheroma progression and regression. J Am Coll Cardiol. 2015;65(13):1273-1282. doi:10.1016/j.jacc.2015.01.036

40. Maron D.J, Hochman J S, Reynolds HR, et al; ISCHEMIA Research Group. Initial invasive or conservative strategy for stable coronary disease. N Engl J Med. 2020;382(15):1395-1407. doi:10.1056/NEJMoa1915922

Issue
Federal Practitioner - 41(1)a
Issue
Federal Practitioner - 41(1)a
Page Number
1
Page Number
1
Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>0124 FED epub ASCVD</fileName> <TBEID>0C02EE32.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EE32</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240109T235211</firstPublished> <LastPublished>20240109T235211</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240109T235211</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Joel Kupfer, MDa; Helme Silvet, MDb; Samuel M. Aguayo, MDa</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Lung cancer is the most common cause of cancer mortality, and cigarette smoking is the most significant risk factor. Several randomized clinical trials have sho</metaDescription> <articlePDF/> <teaserImage/> <title>Preventing ASCVD Events: Using Coronary Artery Calcification Scores to Personalize Risk and Guide Statin Therapy</title> <deck/> <eyebrow>Clinical Review</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2967</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Clinical Review | 2967<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">194</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Preventing ASCVD Events: Using Coronary Artery Calcification Scores to Personalize Risk and Guide Statin Therapy</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Lung cancer is the most common cause of cancer mortality, and cigarette smoking is the most significant risk factor. Among smokers at high risk for lung cancer, atherosclerotic cardiovascular disease (ASCVD) also poses a significant risk for morbidity and mortality. Fortunately, there are opportunities of the prevention of ASCVD events during lung cancer screening (LCS).<br/><br/><b>Observations:</b> Chest low-dose computed tomography (LDCT) scans used for LCS provide information about the absence or severity of coronary artery calcification (CAC), another independent risk factor of ASCVD events. Of note, there are clinically important differences in using CAC scores to guide primary prevention and statin therapy in smokers eligible for LCS compared with those of the general population. This review article focuses on these differences. <b>Conclusions: </b>We provide recommendations on using CAC scores from LDCT to guide the prevention of ASCVD events in LCS in addition to using cardiac testing and when referral to a cardiovascular specialist should be considered.<b> </b></p> <p><span class="Drop">L</span>ung cancer is the most common cause of cancer mortality, and cigarette smoking is the most significant risk factor. Several randomized clinical trials have shown that lung cancer screening (LCS) with nonelectrocardiogram (ECG)-gated low-dose computed tomography (LDCT) reduces both lung cancer and all-cause mortality.<sup>1,2</sup> Hence, the US Preventive Screening Task Force (USPSTF) recommends annual screening with LDCT in adults aged 50 to 80 years who have a 20-pack-year smoking history and currently smoke or have quit within the past 15 years.<sup>3</sup> </p> <p>Smoking is also an independent risk factor for atherosclerotic cardiovascular disease (ASCVD), and LCS clinical trials acknowledge that mortality from ASCVD events exceeds that of lung cancer.<sup>4,5</sup> In an analysis of asymptomatic individuals from the Framingham Heart Offspring study who were eligible for LCS, the ASCVD event rate during a median (IQR) follow-up of 11.4 (9.7-12.0) years was 12.6%.<sup>6</sup> However, despite the high rate of ASCVD events in this population, primary prevention strategies are consistently underused. In a study of 5495 individuals who underwent LCS with LDCT, only 40% of those eligible for statins had one prescribed, underscoring the missed opportunity for preventing ASCVD events during LCS.<sup>7</sup> Yet the interactions for shared decision making and the availability of coronary artery calcification (CAC) scores from the LDCT provide an ideal window for intervening and preventing ASCVD events during LCS.<br/><br/>CAC is a hallmark of atherosclerotic plaque development and is proportional to plaque burden and ASCVD risk.<sup>8</sup> Because of the relationship between CAC, subclinical atherosclerosis, and ASCVD risk, there is an opportunity to use CAC detected by LDCT to predict ASCVD risk and guide recommendations for statin treatment in individuals enrolled in LCS. Traditionally, CAC has been visualized by ECG-gated noncontrast CT scans with imaging protocols specifically designed to visualize the coronary arteries, minimize motion artifacts, and reduce signal noise. These scans are specifically done for primary prevention risk assessment and report an Agatston score, a summed measure based on calcified plaque area and maximal density.<sup>9</sup> Results are reported as an overall CAC score and an age-, sex-, and race-adjusted percentile of CAC. Currently, a CAC score ≥ 100 or above the 75th percentile for age, sex, and race is considered abnormal.<br/><br/>High-quality evidence supports CAC scores as a strong predictor of ASCVD risk independent of age, sex, race, and other traditional risk factors.<sup>10-12</sup> In asymptomatic individuals, a CAC score of 0 is a strong, negative risk factor associated with very low annualized mortality rates and cardiovascular (CV) events, so intermediate-risk individuals can be reclassified to a lower risk group avoiding or delaying statin therapy.<sup>13</sup> As a result, current primary prevention guidelines allow for CAC scoring in asymptomatic, intermediate-risk adults where the clinical benefits of statin therapy are uncertain, knowing the CAC score will aid in the clinical decision to delay or initiate statin therapy.<br/><br/>Unlike traditional ECG-gated CAC scoring, LDCT imaging protocols are non–ECG-gated and performed at variable energy and slice thickness to optimize the detection of lung nodules. Early studies suggested that CAC detected by LDCT could be used in lieu of traditional CAC scoring to personalize risk.<sup>14,15</sup> Recently, multiple studies have validated the accuracy and reproducibility of LDCT to detect and quantify CAC. In both the NELSON and the National Lung Screening Trial (NLST) LCS trials, higher visual and quantitative measures of CAC were independently and incrementally associated with ASCVD risk.<sup>16,17</sup> A subsequent review and meta-analysis of 6 LCS trials confirmed CAC detected by LDCT to be an independent predictor of ASCVD events regardless of the method used to measure CAC.<sup>18<br/><br/></sup>There is now consensus that either an Agatston score or a visual estimate of CAC be reported on all noncontrast, noncardiac chest CT scans irrespective of the indication or technique, including LDCT scans for LCS using a uniform reporting system known as the Coronary Artery Calcium Data and Reporting System (CAC-DRS).<sup>19</sup> The CAC-DRS simplifies reporting and adds modifiers indicating if the reported score is visual (V) or Agatston (A) and number of vessels involved. For example, CAC-DRS A0 or CAC-DRS V0 would indicate an Agatston score of 0 or a visual score of 0. CAC-DRS A1/N2 would indicate a total Agatston score of 1-99 in 2 coronary arteries. The currently agreed-on CAC-DRS risk groups are listed in the Table, along with their corresponding visual score or Agatston score and anticipated 10-year event rate, irrespective of other risk factors.<sup>20</sup>As LCS efforts increase, primary care practitioners will receive LDCT reports that now incorporate an estimation of CAC (visual or quantitative). Thus, it will be increasingly important to know how to interpret and use these scores to guide clinical decisions regarding the initiation of statin therapy, referral for additional testing, and when to seek specialty cardiology care. For instance, does the absence of CAC (CAC = 0) on LDCT predict a low enough risk for statin therapy to be delayed or withdrawn? Does increasing CAC scores on follow-up LDCT in individuals on statin therapy represent treatment failure? When should CAC scores trigger additional testing, such as a stress test or referral to cardiology specialty care? </p> <h2>Primary Prevention in LCS</h2> <p>The initial approach to primary prevention in LCS is no different from that recommended by the 2018 multisociety guidelines on the management of blood cholesterol, the 2019 American College of Cardiology/American Heart Association (ACC/AHA) guideline on primary prevention, or the 2022 USPTSF recommendations on statin use for primary prevention of CV disease in adults.<sup>21-23</sup> For a baseline low-density lipoprotein cholesterol (LDL-C) ≥ 190 mg/dL, high-intensity statin therapy is recommended without further risk stratification. Individuals with diabetes also are at higher-than-average risk, and moderate-intensity statin therapy is recommended.</p> <p>For individuals not in either group, a validated ASCVD risk assessment tool is recommended to estimate baseline risk. The most validated tool for estimating risk in the US population is the 2013 ACC/AHA Pooled Cohort Equation (PCE) which provides an estimate of the 10-year risk for fatal and myocardial infarction and fatal and nonfatal stroke.<sup>24</sup> The PCE risk calculator uses age, presence of diabetes, sex, smoking history, total cholesterol, high-density lipoprotein cholesterol, systolic blood pressure, and treatment for hypertension to place individuals into 1 of 4 risk groups: low (&lt; 5%), borderline (5% to &lt; 7.5%), intermediate (≥ 7.5% to &lt; 20%), and high (≥ 20%). Clinicians should be aware that the PCE only considers current smoking history and not prior smoking history or cumulative pack-year history. This differs from eligibility for LCS where recent smoking plays a larger role. All these risk factors are important to consider when evaluating risk and discussing risk-reducing strategies like statin therapy.<br/><br/>The 2018 multisociety guidelines and the 2019 primary prevention guidelines set the threshold for considering initiation of statin therapy at intermediate risk ≥ 7.5%.<sup>21,22</sup> The 2020 US Department of Veterans Affairs/Department of Defense guidelines set the threshold for considering statin therapy at an estimated 10-year event rate of 12%, whereas the 2022 UPSTF recommendations set the threshold at 10% with additional risk factors as the threshold for statin therapy.<sup>23,25</sup> The reasons for these differences are beyond the scope of this review, but all these guidelines use the PCE to estimate baseline risk as the starting point for clinical decision making. <br/><br/>The PCE was originally derived and validated in population studies dating to the 1960s when the importance of diet, exercise, and smoking cessation in reducing ASCVD events was not well appreciated. The application of the PCE in more contemporary populations shows that it overestimates risk, especially in older individuals and women.<sup>26,27</sup> Overestimation of risk has the potential to result in the initiation of statin therapy in individuals in whom the actual clinical benefit would otherwise be small. <br/><br/>To address this issue, current guidelines allow the use of CAC scoring to refine risk in individuals who are classified as intermediate risk and who otherwise desire to avoid lifelong statin therapy. Using current recommendations, we make suggestions on how to use CAC scores from LDCT to aid in clinical decision making for individuals in LCS (Figure). </p> <h3>No Coronary Artery Calcification</h3> <p>Between 25% and 30% of LDCT done for LCS will show no CAC.<sup>14,16</sup> In general population studies, a CAC score of 0 is a strong negative predictor when there are no other risk factors.<sup>13,28</sup> In contrast, the negative predictive ability of a CAC score of 0 in individuals with a smoking history who are eligible for LCS is unproven. In multivariate modeling, a CAC score of 0 did not reduce the significant hazard of all-cause mortality in patients with diabetes or smokers.<sup>29</sup> In an analysis of 44,042 individuals without known heart disease referred for CAC scoring, the frequency of a CAC score of 0 was only modestly lower in smokers (38%) compared with nonsmokers (42%), yet the all-cause mortality rate was significantly higher.<sup>30</sup> In addition, Multi-Ethnic Study of Atherosclerosis (MESA) participants who were current smokers or eligible for LCS and had a CAC score of 0 had an observed 11-year ASCVD event rate of 13.4% and 20.8%, respectively, leading to the conclusion that a CAC score of 0 may not be predictive of minimal risk in smokers and those eligible for LCS.<sup>31</sup> Additionally, in LCS-eligible individuals, the PCE underestimated event rates and incorporation of CAC scores did not significantly improve risk estimation. Finally, data from the NLST screening trial showed that the absence of CAC on LDCT was not associated with better survival or lower CV mortality compared with individuals with low CAC scores.<sup>32</sup></p> <p>The question of whether individuals undergoing LCS with LDCT who have no detectable CAC can avoid statin therapy is an unresolved issue; no contemporary studies have looked specifically at the relationship between estimated risk, a CAC score of 0, and ASCVD outcomes in individuals participating in LCS. For these reasons, we recommend moderate-intensity statin therapy when the estimated risk is intermediate because it is unclear that either an Agatston score of 0 reclassifies intermediate-risk LCS-eligible individuals to a lower risk group. <br/><br/>For the few borderline risk (estimated risk, 5% to &lt; 7.5%) LCS-eligible individuals, a CAC score of 0 might confer low short-term risk but the long-term benefit of statin therapy on reducing subsequent risk, the presence of other risk factors, and the willingness to stop smoking should all be considered. For these individuals who elect to avoid statin therapy, annual re-estimation of risk at the time of repeat LDCT is recommended. In these circumstances, referral for traditional Agatston scoring is not likely to change decision making because the sensitivity of the 2 techniques is very similar. </p> <h3>Agatston Score of 1-99 or CAC-DRS or Visual Score of 1</h3> <p>In general population studies, these scores correspond to borderline risk and an estimated 10-year event rate of just under 7.5%.<sup>20</sup> In both the NELSON and NLST LCS trials, even low amounts of CAC regardless of the scoring method were associated with higher observed ASCVD mortality when adjusted for other baseline risk factors.<sup>32</sup> Thus, in patients undergoing LCS with intermediate and borderline risk, a CAC score between 1 and 99 or a visual estimate of 1 indicates the presence of subclinical atherosclerosis, and moderate-intensity statin therapy is reasonable.</p> <h3>Agatston Score of 100-299 or CAC-DRS or Visual Score of 2</h3> <p>Across all ages, races, and sexes, CAC scores between 100 to 299 are associated with an event rate of about 15% over 10 years.<sup>20</sup> In the NELSON LCS trial, the adjusted hazard ratio for ASCVD events with a nontraditional Agatston score of 101 to 400 was 6.58.<sup>33</sup> Thus, in patients undergoing LCS with a CAC score of 100 to 299, regardless of the baseline risk estimate, the projected absolute event rate at 10 years would be about 20%. Moderate-intensity statin therapy is recommended to reduce the baseline LDL-C by 30% to 49%.</p> <h3>Agatston Score of &gt; 300 orCAC-DRS or Visual Score of 3</h3> <p>Agatston CAC scores &gt; 300 are consistent with a 10-year incidence of ASCVD events of &gt; 15% regardless of age, sex, or race and ethnicity.<sup>20</sup> In the Calcium Consortium, a CAC &gt; 400 was correlated with an event rate of 13.6 events/1000 person-years.<sup>12</sup> In a Walter Reed Military Medical Center study, a CAC score &gt; 400 projected a cumulative incidence of ASCVD events of nearly 20% at 10 years.<sup>34</sup> In smokers eligible for LCS, a CAC score &gt; 300 projected a 10-year ASCVD event rate of 25%.<sup>29</sup> In these patients, moderate-intensity statin therapy is recommended, although high-intensity statin therapy can be considered if there are other risk factors. </p> <h3>Agatston Score ≥ 1000</h3> <p>The 2018 consensus statement on CAC reporting categorizes all CAC scores &gt; 300 into a single risk group because the recommended treatment options do not differ.<sup>19</sup> However, recent data suggest this might not be the case since individuals with very high CAC scores experience high rates of events that might justify more aggressive intervention. In an analysis of individuals who participated in the CAC Consortium with a CAC score ≥ 1000, the all-cause mortality rate was 18.8 per 1000 person-years with a CV mortality rate of 8 per 1000 person-years.<sup>35</sup> Individuals with very high levels of CAC &gt; 1000 also have a greater number of diseased coronary arteries, higher involvement of the left main coronary artery, and significantly higher event rates compared with those with a CAC of 400 to 999.<sup>36</sup> In an analysis of individuals from the NLST trial, nontraditionally measured Agatston score &gt; 1000 was associated with a hazard ratio for coronary artery disease (CAD) mortality of 3.66 in men and 5.81 in women.<sup>17</sup> These observed and projected levels of risk are like that seen in secondary prevention trials, and some experts have recommended the use of high-intensity statin therapy to reduce LDL-C to &lt; 70 mg/dL.<sup>37</sup></p> <h2>Primary Prevention in Individuals aged 76 to 80 years </h2> <p>LCS can continue through age 80 years, while the PCE and primary prevention guidelines are truncated at age 75 years. Because age is a major contributor to risk, many of these individuals will already be in the intermediate- to high-risk group. However, the net clinical benefit of statin therapy for primary prevention in this age group is not well established, and the few primary prevention trials in this group have not demonstrated net clinical benefit.<sup>38</sup> As a result, current guidelines do not provide specific treatment recommendations for individuals aged &gt; 75 years but recognize the value of shared decision making considering associated comorbidities, age-related risks of statin therapy, and the desires of the individual to avoid ASCVD-related events even if the net clinical benefit is low. </p> <p>Older individuals with elevated CAC scores should be informed about the risk of ASCVD events and the potential but unproven benefit of moderate-intensity statin therapy. Older individuals with a CAC score of 0 likely have low short-term risk of ASCVD events and withholding statin therapy is not unreasonable.</p> <h3>CAC Scores on Annual LDCT Scans </h3> <p>Because LCS requires annual LDCT scans, primary care practitioners and patients need to understand the significance of changing CAC scores over time. For individuals not on statin therapy, increasing calcification is a marker of progression of subclinical atherosclerosis. Patients undergoing LCS not on statin who have progressive increases in their CAC should consider initiating statin therapy. Individuals who opted not to initiate statin therapy who subsequently develop CAC should be re-engaged in a discussion about the significance of the finding and the clinically proven benefits of statin therapy in individuals with subclinical atherosclerosis. These considerations do not apply to individuals already on statin therapy. Statins convert lipid-rich plaques to lipid-depleted plaques, resulting in increasing calcification. As a result, CAC scores do not decrease and may increase with statin therapy.<sup>39</sup> Individuals participating in annual LCS should be informed of this possibility. Also, in these individuals, referral to specialty care as a treatment failure is not supported by the literature.</p> <p>Furthermore, serial CAC scoring to titrate the intensity of statin therapy is not currently recommended. The goal with moderate-intensity statin therapy is a 30% to 49% reduction from baseline LDL-C. If this milestone is not achieved, the statin dose can be escalated. For high-intensity statin therapy, the goal is a &gt; 50% reduction. If this milestone is not achieved, then additional lipid-lowering agents, such as ezetimibe, can be added.</p> <h3>Further ASCVD Testing</h3> <p>LCS with LDCT is associated with improved health outcomes, and LDCT is the preferred imaging modality. The ability of LDCT to detect and quantify CAC is sufficient for clinical decision making. Therefore, obtaining a traditional CAC score increases radiation exposure without additional clinical benefits.</p> <p>Furthermore, although referral for additional testing in those with nonzero CAC scores is common, current evidence does not support this practice in asymptomatic individuals. Indeed, the risks of LCS include overdiagnosis, excessive testing, and overtreatment secondary to the discovery of other findings, such as benign pulmonary nodules and CAC. With respect to CAD, randomized controlled trials do not support a strategy of coronary angiography and intervention in asymptomatic individuals, even with moderate-to-severe ischemia on functional testing.<sup>40</sup> As a result, routine stress tests to diagnose CAD or to confirm the results of CAC scores in asymptomatic individuals are not recommended. The only potential exception would be in select cases where the CAC score is &gt; 1000 and when calcium is predominately located in the left main coronary artery.</p> <h2>Conclusions</h2> <p>LCS provides smokers at risk for lung cancer with the best probability to survive that diagnosis, and coincidentally LCS may also provide the best opportunity to prevent ASCVD events and mortality. Before initiating LCS, clinicians should initiate a shared decision making conversation about the benefits and risks of LDCT scans. In addition to relevant education about smoking, during shared decision making, the initial ASCVD risk estimate should be done using the PCE and when appropriate the benefits of statin therapy discussed. Individuals also should be informed of the potential for identifying CAC and counseled on its significance and how it might influence the decision to recommend statin therapy. </p> <p>In patients undergoing LCS with an estimated risk of ≥<b> </b>7.5% to &lt; 20%, moderate-intensity statin therapy is indicated. In this setting, a CAC score &gt; 0 indicates subclinical atherosclerosis and should be used to help direct patients toward initiating statin therapy. Unfortunately, in patients undergoing LCS a CAC score of 0 might not provide protection against ASCVD, and until there is more information to the contrary, these individuals should at least participate in shared decision making about the long-term benefits of statin therapy in reducing ASCVD risk. Because LDCT scanning is done annually, there are opportunities to review the importance of prevention and to adjust therapy as needed to achieve the greatest reduction in ASCVD. Reported elevated CAC scores on LDCT provide an opportunity to re-engage the patient in the discussion about the benefits of statin therapy if they are not already on a statin, or consideration for high-intensity statin if the CAC score is &gt; 1000 or reduction in baseline LDL-C is &lt; 30% on the current statin dose.</p> <h3> Author affiliations </h3> <p> <em><sup>a</sup>Carl T. Hayden Veterans Affairs Medical Center, Phoenix, Arizona<sup>b</sup>Veterans Affairs Loma Linda Healthcare System, California</em> </p> <h3> Author disclosures </h3> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <h3> Disclaimer </h3> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner,</i> Frontline Medical Communications Inc., the U.S. Government, or any of its agencies.</em> </p> <h3> References </h3> <p class="reference"> 1. de Koning HJ, van der Aalst CM, Oudkerk M. Lung-cancer screening and the NELSON Trial. Reply. <i>N Engl J Med. </i>2020;382(22):2165-2166. doi:10.1056/NEJMc2004224<br/><br/> 2. Aberle T, Adams DR, Berg AM, et al; National Lung Screening Trial Research Team. Reduced lung-cancer mortality with low-dose computed tomographic screening. <i>N Engl J Med. </i>2011;365(5):396-409. doi:10.1056/NEJMoa1102873<br/><br/> 3. Krist AH, Davidson KW, Mangione CM, et al; US Preventive Services Task Force. Screening for lung cancer: US Preventive Services Task Force recommendation statement. <i>JAMA. </i>2021;25(10):962-970. doi:10.1001/jama.2021.1117<br/><br/> 4. Jha P, Ramasundarahettige C, Landsman V. 21st-century hazards of smoking and benefits of cessation in the United States. <i>N Engl J Med</i>. 2013;368(4):341-350. doi:10.1056/NEJMsa1211128<br/><br/> 5. Khan SS, Ning H, Sinha A, et al. Cigarette smoking and competing risks for fatal and nonfatal cardiovascular disease subtypes across the life course. <i>J Am Heart Assoc.</i> 2021;10(23):e021751. doi:10.1161/JAHA.121.021751<br/><br/> 6. Lu MT, Onuma OK, Massaro JM, et al. Lung cancer screening eligibility in the community: cardiovascular risk factors, coronary artery calcification, and cardiovascular events. <i>Circulation</i>. 2016;134(12):897-899. doi:10.1161/CIRCULATIONAHA.116.023957<br/><br/> 7. Tailor TD, Chiles C, Yeboah J, et al. Cardiovascular risk in the lung cancer screening population: a multicenter study evaluating the association between coronary artery calcification and preventive statin prescription. <i>J Am Coll Radiol. </i>2021;18(9):1258-1266. doi:10.1016/j.jacr.2021.01.015<br/><br/> 8. Mori H, Torii S, Kutyna M, et al. Coronary artery calcification and its progression: what does it really mean? <i>JACC Cardiovasc Imaging</i>. 2018;11(1):127-142. doi:10.1016/j.jcmg.2017.10.012</p> <p class="reference">10. Nasir K, Bittencourt MS, Blaha MJ, et al. Implications of coronary artery calcium testing among statin candidates according to American College of Cardiology/American Heart Association cholesterol management guidelines: MESA (Multi-Ethnic Study of Atherosclerosis). <i>J Am Coll Cardiol</i>. 2015;66(15): 1657-1668. doi:10.1016/j.jacc.2015.07.066 </p> <p class="reference">11. Detrano R, Guerci AD, Carr JJ, et al. Coronary calcium as a predictor of coronary events in four racial or ethnic groups. <i>N Engl J Med. </i>2008;358(13):1336-1345. doi:10.1056/NEJMoa072100 <br/><br/>12. Grandhi GR, Mirbolouk M, Dardari ZA. Interplay of coronary artery calcium and risk factors for predicting CVD/CHD Mortality: the CAC Consortium. <i>JACC Cardiovasc Imaging. </i>2020;13(5):1175-1186. doi:10.1016/j.jcmg.2019.08.024<br/><br/>13. Blaha M, Budoff MJ, Shaw J. Absence of coronary artery calcification and all-cause mortality. <i>JACC Cardiovasc Imaging. </i>2009;2(6):692-700. doi:10.1016/j.jcmg.2009.03.009<br/><br/>14. Shemesh J, Henschke CI, Farooqi A, et al. Frequency of coronary artery calcification on low-dose computed tomography screening for lung cancer. <i>Clin Imaging</i>. 2006;30(3):181-185. doi:10.1016/j.clinimag.2005.11.002<br/><br/>15. Shemesh J, Henschke C, Shaham D, et al. Ordinal scoring of coronary artery calcifications on low-dose CT scans of the chest is predictive of death from cardiovascular disease. <i>Radiology</i>. 2010;257:541-548. doi:10.1148/radiol.10100383<br/><br/>16. Jacobs PC, Gondrie MJ, van der Graaf Y, et al. Coronary artery calcium can predict all-cause mortality and cardiovascular events on low-dose CT screening for lung cancer. <i>AJR Am J Roentgenol</i>. 2012;198(3):505-511. doi:10.2214/AJR.10.5577<br/><br/>17. Lessmann N, de Jong PA, Celeng C, et al. Sex differences in coronary artery and thoracic aorta calcification and their association with cardiovascular mortality in heavy smokers. <i>JACC Cardiovasc Imaging</i>.<i> </i>2019;12(9):1808-1817. doi:10.1016/j.jcmg.2018.10.026<br/><br/>18. Gendarme S, Goussault H, Assie JB, et al. Impact on all-cause and cardiovascular mortality rates of coronary artery calcifications detected during organized, low-dose, computed-tomography screening for lung cancer: systematic literature review and meta-analysis. <i>Cancers (Basel)</i>. 2021;13(7):1553. doi:10.3390/cancers13071553<br/><br/>19. Hecht HS, Blaha MJ, Kazerooni EA, et al. CAC-DRS: coronary artery calcium data and reporting system. An expert consensus document of the Society of Cardiovascular Computed Tomography (SCCT). <i>J Cardiovasc Comput Tomogr. </i>2018;12(3):185-191. doi:10.1016/j.jcct.2018.03.008<br/><br/>20. Budoff MJ, Young R, Burke G, et al. Ten-year association of coronary artery calcium with atherosclerotic cardiovascular disease (ASCVD) events: the multi-ethnic study of atherosclerosis (MESA). <i>Eur Heart J. </i>2018;39(25):2401-2408. doi:10.1093/eurheartj/ehy217<br/><br/>21. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. <i>Circulation</i>. 2019;139(25):e1046-e1081. doi:10.1161/CIR.0000000000000624<br/><br/>22. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. <i>Circulation. </i>2019;140(11):e596-e646. doi:10.1161/CIR.0000000000000678<br/><br/>23. Mangione CM, Barry MJ, Nicholson WK, et al; US Preventive Services Task Force. Statin use for the primary prevention of cardiovascular disease in adults: US Preventive Services Task Force recommendation statement. <i>JAMA. </i>2022;328(8):746-753. doi:10.1001/jama.2022.13044<br/><br/>24. Stone NJ, Robinson JG, Lichtenstein AH, et al; American College of Cardiology/American Heart Association Task Force on Practice. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. <i>J Am Coll Cardiol. </i>2014;63(25 pt B):2889-2934. doi:10.1016/j.jacc.2013.11.002</p> <p class="reference">25. US Department of Veterans Affairs, Department of Defense. VA/DoD clinical practice guideline. Updated August 25, 2021. Accessed November 3, 2023. https://www.healthquality.va.gov/guidelines/cd/lipids <br/><br/>26. DeFilippis AP, Young, R, Carrubba CJ, et al. An analysis of calibration and discrimination among multiple cardiovascular risk scores in a modern multiethnic cohort. <i>Ann Intern Med. </i>2015;162(4):266-275. doi:10.7326/M14-1281<br/><br/>27. Rana JS, Tabada GH, Solomon, MD, et al. Accuracy of the atherosclerotic cardiovascular risk equation in a large contemporary, multiethnic population. <i>J Am Coll Cardiol</i>. 2016;67(18):2118-2130. doi:10.1016/j.jacc.2016.02.055<br/><br/>28. Sarwar A, Shaw LJ, Shapiro MD, et al. Diagnostic and prognostic value of absence of coronary artery calcification. <i>JACC Cardiovasc Imaging.</i> 2009;2(6):675-688. doi:10.1016/j.jcmg.2008.12.031<br/><br/>29. McEvoy JW, Blaha MJ, Rivera JJ, et al. Mortality rates in smokers and nonsmokers in the presence or absence of coronary artery calcification. <i>JACC Cardiovasc Imaging</i>. 2012;5(10):1037-1045. doi:10.1016/j.jcmg.2012.02.017<br/><br/>30. Leigh A, McEvoy JW, Garg P, et al. Coronary artery calcium scores and atherosclerotic cardiovascular disease risk stratification in smokers. <i>JACC Cardiovasc Imaging</i>. 2019;12(5):852-861. doi:10.1016/j.jcmg.2017.12.017<br/><br/>31. Garg PK, Jorgensen NW, McClelland RL, et al. Use of coronary artery calcium testing to improve coronary heart disease risk assessment in lung cancer screening population: The Multi-Ethnic Study of Atherosclerosis (MESA). <i>J Cardiovasc Comput Tomagr</i>. 2018;12(6):439-400.<br/><br/>32. Chiles C, Duan F, Gladish GW, et al. Association of coronary artery calcification and mortality in the national lung screening trial: a comparison of three scoring methods. <i>Radiology.</i> 2015;276(1):82-90. doi:10.1148/radiol.15142062<br/><br/>33. Takx RA, Isgum I, Willemink MJ, et al. Quantification of coronary artery calcium in nongated CT to predict cardiovascular events in male lung cancer screening participants: results of the NELSON study. <i>J Cardiovasc Comput Tomogr. </i>2015;9(1):50-57. doi:10.1016/j.jcct.2014.11.006<br/><br/>34. Mitchell JD, Paisley R, Moon P, et al. Coronary artery calcium and long-term risk of death, myocardial infarction, and stroke: The Walter Reed Cohort Study. <i>JACC Cardiovasc Imaging.</i> 2018;11(12):1799-1806. doi:10.1016/j.jcmg.2017.09.003<br/><br/>35. Peng AW, Mirbolouk M, Orimoloye OA, et al. Long-term all-cause and cause-specific mortality in asymptomatic patients with CAC &gt;/=1,000: results from the CAC Consortium. <i>JACC Cardiovasc Imaging. </i>2019;13(1, pt 1):83-93. doi:10.1016/j.jcmg.2019.02.005 <br/><br/>36. Peng AW, Dardari ZA. Blumenthal RS, et al. Very high coronary artery calcium (&gt;/=1000) and association with cardiovascular disease events, non-cardiovascular disease outcomes, and mortality: results from MESA. <i>Circulation</i>. 2021;143(16):1571-1583. doi:10.1161/CIRCULATIONAHA.120.050545<br/><br/>37. Orringer CE, Blaha MJ, Blankstein R, et al. The National Lipid Association scientific statement on coronary artery calcium scoring to guide preventive strategies for ASCVD risk reduction. <i>J Clin Lipidol</i>. 2021;15(1):33-60. doi:10.1016/j.jacl.2020.12.005</p> <p class="reference">38. Sheperd J, Blauw GJ, Murphy MB, et al; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk. Pravastatin in elderly individuals at risk of vascular disease. (PROSPER): a randomized controlled trial. <i>Lancet</i>. 2002;360:1623-1630. doi:10.1016/s0140-6736(02)11600-x</p> <p class="reference">39. Puri R, Nicholls SJ, Shao M, et al. Impact of statins on serial coronary calcification during atheroma progression and regression. <i>J Am Coll Cardiol</i>. 2015;65(13):1273-1282. doi:10.1016/j.jacc.2015.01.036</p> <p class="reference">40. Maron D.J, Hochman J S, Reynolds HR, et al; ISCHEMIA Research Group. Initial invasive or conservative strategy for stable coronary disease. <i>N Engl J Med</i>. 2020;382(15):1395-1407. doi:10.1056/NEJMoa1915922</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Axillary Contact Dermatitis: An Update on Potential Allergens and Management

Article Type
Changed
Tue, 01/09/2024 - 12:49
Display Headline
Axillary Contact Dermatitis: An Update on Potential Allergens and Management

Approximately 20% of the general population has a contact allergy.1 Allergic contact dermatitis (ACD) is a delayed type IV hypersensitivity reaction mediated by T lymphocytes.2 Axillary ACD presentation is variable but typically includes an eczematous eruption with erythematous scaly patches or plaques. Common products in contact with the axillae include deodorants, antiperspirants, razors, bodywash, and clothing.

Axillary skin is distinct from skin elsewhere on the body due to both anatomical characteristics and unique human self-care practices. Axillary skin has reduced barrier function, faster stratum corneum turnover, and altered lipid levels.3-5 Moreover, the axillae often are subject to shaving or other hair removal practices that alter the local environment, as layers of stratum corneum and hair are mechanically removed, which causes irritation and predisposes the skin to enhanced sensitivity to topical exposures.6,7 With the abundance of apocrine and eccrine glands, the axillae are prone to sweat, which also can accentuate contact allergy.2,3 Other factors, such as occlusion and friction, contribute to axillary contact allergy.8,9

Patch testing is the gold standard for the diagnosis of ACD and aids in identification of culprit allergens. A thorough patient history and examination of the rash distribution may provide further clues; for example, dermatitis due to a deodorant typically affects the vault, whereas textile dye dermatitis tends to spare the vault.10,11 Baseline-limited patch testing detects up to two-thirds of clinically relevant allergens.12 Therefore, patients may require subsequent testing with supplemental allergens.

The differential diagnosis for axillary lesions is broad—including inflammatory diseases such as irritant contact dermatitis and hidradenitis suppurativa, genetic disorders such as Hailey-Hailey disease, and infectious causes such as erythrasma—but may be narrowed with a thorough physical examination and patient history, histopathology, bedside diagnostic techniques (eg, scrapings and Wood lamp examination), and patch testing. Systemic contact dermatitis (SCD) or symmetrical drug-related intertriginous and flexural exanthema (SDRIFE) also may be suspected in cases of intertriginous dermatoses.

We review the potential allergens in products used on the axillae as well as the management of axillary ACD. We also discuss axillary dermatitis as a manifestation of SCD and SDRIFE.

Top Allergens in Products Used on the Axillae

Fragrance—A 1982 North American Contact Dermatitis Group study on cosmetic products identified fragrances as the most common cause of ACD,13 and this trend continues to hold true with more recent data.14 The incidence of fragrance allergy may be increasing, with positive patch tests to a fragrance chemical in 10% of patch test clinic populations.15 Fragrances are a ubiquitous ingredient in deodorants and antiperspirants, which are important sources implicated in the development and elicitation of fragrance ACD.16 One study found that fragrance was present in 97 of 107 (90%) deodorants available at Walgreens pharmacies.11

In a study of patients with a history of an axillary rash caused by a deodorant spray, Johansen et al17 reported that the likelihood of fragrance allergy is increased by a factor of 2.4. This risk of developing a fragrance allergy may be exacerbated in those who shave; Edman18 reported that the odds ratio of developing a fragrance allergy among men who shave their beards was 2.9. Although there are no specific data on the effects of shaving on ACD, shaving in general can induce localized irritation and increase percutaneous absorption.19

 

 

The individual fragrance components in deodorants most likely to cause ACD include hydroxycitronellal, eugenol, and geraniol—all constituent ingredients in patch test formulations of fragrance mixture I.11,20 Other common fragrance allergens associated with ACD include hydroxymethylpentylcyclohexenecarboxaldehyde, farnesol, and balsam of Peru.21-27 Hydroperoxides of limonene and linalool, common fragrances in detergents and personal care products, are increasingly recognized as contact allergens and have been reported to cause axillary ACD from deodorants.28-30

Dermatitis involving the bilateral axillary vaults wherever deodorant or antiperspirant was directly applied is the most common presentation of ACD due to fragrance (Figure 1).17 An eczematous eruption is common, though scale may be less apparent than in nonflexural regions. Axillary ACD secondary to fragrances also may result from use of fragranced laundry detergents, fabric softeners, soaps, and perfumes, and may spare the vaults.10,29,31,32 Less common presentations of axillary ACD due to fragrance include pigmented dermatoses; for example, ACD from an antiperspirant containing hydroperoxide of limonene presented as hyperpigmented patches with minimal erythema and scaling in the edges of the axillary folds.33,34

Musicante_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Allergic%20contact%20dermatitis%20of%20the%20axillary%20vault%20secondary%20to%20use%20of%20scented%20antiperspirant%2Fdeodorant%20in%20a%20patient%20with%20positive%20patch%20test%20results%20to%20propylene%20glycol%2C%20balsam%20of%20Peru%2C%20and%20quaternium-15.%3C%2Fp%3E

Diagnosis of a fragrance ACD typically is made with a standard patch test series including fragrance mixture I and balsam of Peru, which may detect 75% and 50% of fragrance sensitivities, respectively.35 Patch testing may be followed with a repeated open application test of the product in question.36 Additionally, it may be appropriate to test for other fragrance allergens including balsam of Tolu, fragrance mixture II, lichen acid mix, and hydroxyperoxides of linalool and limonene (among other botanicals) if standard patch testing is negative and suspicion of fragrance ACD remains elevated.11

Propylene Glycol—Propylene glycol (PG)—a versatile substance that functions as a solvent, humectant, emulsifier, stabilizer, and antimicrobial—is the second most common contact allergen present in deodorants.11 It is prevalent in both personal care and household products, including deodorants, cosmetics, foods, toothpaste, cleaning agents, and detergents.11,37 Propylene glycol is both an allergen and an irritant. Among deodorants/antiperspirants, PG is both a common irritant and allergen, as its concentration may be particularly high (as much as 73%).38 One commonly reported example of PG contact dermatitis is from the topical medicament minoxidil.39,40

Patch testing data have demonstrated a positivity rate for PG ranging between 0.1% to 3.8%. The variability in these findings likely is due to differences in the tested concentrations of PG, as higher concentrations sometimes required to elicit an allergic reaction also may create a stronger irritation effect.41 Propylene glycol irritancy and the occlusive nature of the axillae may enhance sensitization to other allergens, as demonstrated by Agren-Jonsson and Magnusson,42 who reported sensitization to propantheline bromide and trichlorocarbanilide in patients who used a lotion with 90% PG. Many PG-containing products beyond deodorants/antiperspirants may be applied to the axillae, including steroid creams, lotions, shaving creams, and bodywashes.38,43

The diagnosis of PG allergy via patch testing is challenging and at times controversial given its irritant nature. False-positive irritant reactions have been documented, characterized by a weak reaction at 48 hours that is absent by 96 hours (decrescendo reaction). A reaction may not appear until 96 hours (crescendo reaction), which typically indicates a true contact allergy but in the case of PG also may be the substance acting as a “late irritant.”44 Fast (<24 hours) and well-demarcated reactions suggest irritation.45 Regardless, reactions to PG on patch testing, even those regarded as weak, may be considered relevant in consideration of the clinical context.37

Aluminum—Aluminum is the active ingredient in most antiperspirants, typically in the form of aluminum chloride, aluminum chlorohydrate, aluminum zirconium trichlorohydrex gly, or aluminum zirconium tetrachlorohydrex gly.46 Aluminum mechanically obstructs the eccrine glands to reduce sweat.47 Although aluminum is an uncommon allergen, a possible presentation of aluminum allergy is axillary vault dermatitis secondary to antiperspirant use.46 Another potential manifestation is a ringlike reaction to the Finn Chambers (SmartPractice) used in patch testing.46 In one case of aluminum-induced axillary dermatitis, a 28-year-old woman presented with eczema of the axillae, and subsequent patch testing revealed an allergy to aluminum chloride. The rash resolved upon cessation of use of an aluminum-containing deodorant.48

 

 

Aluminum has been reported to cause granulomatous dermatitis in the axillae. This reaction typically presents as red-brown, pruritic papules limited to the area in which deodorant was applied, with histopathology revealing epithelioid granulomas.49-51

Alum deodorants—considered a natural alternative—contain aluminum bound to potassium or ammonium in the form of a crystal or powder. Alum crystal deodorants have been reported to cause both a typical erythematous pruritic dermatitis as well as a granulomatous dermatitis with red-brown papules.52,53 The granulomatous dermatitis caused by either form of aluminum resolves with avoidance and use of topical steroids or topical tacrolimus.49,50,52,53

The diagnosis of aluminum ACD via patch testing may be identified with empty Finn Chambers, which are metallic aluminum, or with patch placement of aluminum chloride hexahydrate, though the former is only positive in patients with a strong allergy.54,55 In 2022, aluminum was named Allergen of the Year by the American Contact Dermatitis Society, with recommendations to conduct patch testing with aluminum chloride hexahydrate 10% rather than the traditional 2% to increase diagnostic yield.55 Additionally, it is recommended that aluminum be included in baseline patch testing for children due to the high prevalence of aluminum allergy in children and early exposure via childhood vaccines.54-56 In patients with aluminum allergy, providers may suggest purchasing aluminum-free deodorants or provide recipes for homemade deodorant that includes ingredients such as arrowroot powder, cornstarch, and diatomaceous earth.46

Nickel—Nickel is the most commonly identified contact allergen on patch testing yet an infrequent cause of axillary dermatitis. A case report from 2014 described axillary dermatitis in a woman that worsened during a positive patch test to nickel. Improvement was noted when the patient switched to titanium shaving razors.57 Nickel allergy also may present in the form of SCD. In one report, a woman developed dermatitis of the flexural areas, including the axillae, 3 months after undergoing a sterilization procedure in which nickel-containing tubal implants were placed.58 Patch testing revealed a positive reaction to nickel. The patient experienced complete resolution of the steroid-resistant dermatitis following removal of the implants via salpingectomy.58

Musicante_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Textile%20allergic%20contact%20dermatitis%20secondary%20to%20a%20deeply%20dyed%20blue%20sweater%20in%20a%20patient%20with%20positive%20patch%20test%20results%20to%20disperse%20blue%20106%2C%20disperse%20blue%20124%2C%20textile%20dye%20mix%2C%20formaldehyde%2C%20and%20methyldibromo%20glutaronitrile%2C%20among%20other%20allergens.%20The%20dermatitis%20involved%20the%20bilateral%20axillary%20rim%20and%20spared%20the%20vault.%3C%2Fp%3E

Textile Dye—In contrast to dermatitis caused by deodorants/antiperspirants, contact allergy to textile dyes presents as dermatitis involving the axillary borders but sparing the axillary vaults (Figures 2 and 3).10 Other potential presentations of textile dye dermatitis include erythema multiforme–like eruptions and erythematous wheal–type reactions.59 Textile dyes are classified as disperse vs nondisperse, with the majority of contact dermatoses caused by disperse dyes, specifically Disperse Orange 1, blue 106, and blue 124.60-62 Ryberg et al61 found that the axilla is one of the more common locations to be affected by textile dye allergy, particularly in women, which was further supported by Seidenari et al,63 who found that skin folds were affected in 27% of study participants allergic to textile dyes (N=437), a finding that is likely due to friction, sweat, and occlusion.62 In one case report of a patient with dermatitis caused by reactive dyes, the garment required 3 washes before the patient experienced resolution of dermatitis.64 For patients with textile dye dermatitis, mitigation strategies include washing clothing before wearing, especially for darkly dyed items; avoiding tight clothing; wearing garments made of cotton, wool, silk, or linen; and choosing light-colored garments.9,64,65

Musicante_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Allergic%20contact%20dermatitis%20of%20the%20axilla%20suspected%20to%20be%20secondary%20to%20black%20textile%20dyes.%20The%20dermatitis%20resolved%20completely%20with%20avoidance%20of%20tightly%20fitted%20black%20clothing.%3C%2Fp%3E

Axillary Dermatitis as a Manifestation of SCD and SDRIFE

Systemic contact dermatitis occurs when an individual who was previously sensitized to a particular allergen develops ACD of the skin with systemic exposure to that allergen or immunochemically related allergens. Exposure may occur via ingestion, inhalation, intravenous, intramuscular, and transepidermal routes.66 Systemic contact dermatitis manifests in a variety of ways, including focal flares at sites of prior contact dermatitis (recall reaction), vesicular hand dermatitis, intertriginous eruptions including axillary dermatitis, and generalized eruptions.67

Systemic contact dermatitis rarely involves systemic symptoms, and onset typically is within days of exposure. The 3 most common groups of allergens causing SCD are metals, medications, and plants and herbals.68 These allergens have all been reported to cause axillary dermatitis via SCD.58,69,70 Foods containing balsam of Peru that may lead to SCD include citrus, chocolate, tomato, and certain alcohols.70,71 Patients with a positive patch test to balsam of Peru may experience improvement of their dermatitis after reduction of balsam of Peru–rich foods from their diet.70 Metals implicated in SCD include mercury, nickel, and gold.72-74 Finally, PG ingestion also has been implicated in cases of SCD.37

 

 

Symmetrical drug-related intertriginous and flexural exanthema is another condition that presents as intertriginous dermatitis and differs from SCD in that the eruption does not require presensitization; there may be no known prior exposure to the agent causing dermatitis. Historically, SDRIFE was described as baboon syndrome because of its frequent involvement of the buttocks with diffuse, well-demarcated, erythematous dermatitis resembling that of a baboon. This term is no longer used due to its insensitive nature and incomplete depiction of SDRIFE, which can affect body sites other than the buttocks.68,75,76 Specific criteria to make this diagnosis include sharply demarcated and/or V-shaped erythema of the gluteal/perianal area, involvement of at least 1 other intertriginous or flexural region, symmetry of affected areas, and an absence of systemic symptoms.76 There also may be papules, pustules, and vesicles present in affected areas. Symmetrical drug-related intertriginous and flexural exanthema most often is caused by β-lactam antibiotics, but other associated drugs include chemotherapeutic agents, such as mitomycin C.76

Histopathology of both SCD and SDRIFE is variable and typically nonspecific, often revealing epidermal spongiosis and a perivascular mononuclear cell infiltrate with occasional neutrophils and eosinophils.76 A case of SCD to mercury presenting as intertriginous dermatitis demonstrated a leukocytoclastic vasculitis pattern on biopsy.77

Systemic contact dermatitis is diagnosed via a patch test, while SDRIFE typically has a negative patch test result and requires oral rechallenge testing, which reproduces the rash within hours.78,79

Additional Allergens Causing Axillary ACD

Although fragrance is the most common allergen in deodorants, other ingredients have been shown to cause axillary ACD (Table).80-90 In addition to these ingredients, allergens not previously mentioned that may be present in deodorants include lanolin, essential oils, and parabens.11 Methylisothiazolinone in laundry detergent also has been found to instigate ACD.91 Fragrances and preservatives in laundry detergents also may contribute to dermatitis.92

CT113001035_Table.jpg

Other products that have caused axillary contact dermatitis include topical exposure to medicaments including clindamycin,93 ethylenediamine in nystatin cream,94 methylprednisolone acetate95 and dipropylene glycol in a hydrocortisone lotion,96 wood dusts from tropical hardwoods,97 and tobacco.98

Management of ACD

The most effective strategy in the management of patients with contact dermatitis is avoidance of the offending agent. Additionally, clinicians may recommend the use of topical steroids and/or calcineurin inhibitors to hasten resolution.2

For patients with contact dermatitis, a clinician may recommend product substitutions with few potential allergens to use prior to patch testing. Patients with a fragrance allergy should look for products specifically labeled as “fragrance free” rather than “hypoallergenic” or “unscented,” as the latter two may still contain minimal amounts of fragrance.35 Patients should be educated on the functions of the allergens to which they are allergic so they may adequately avoid potential sources of contact.99 For suspected textile dye dermatitis, instructing patients to wash clothing before wearing and to avoid synthetic fabrics, dark dyes, and tightly fitted clothing may help.9,64,65

 

 

Differential Diagnosis

The differential diagnosis for axillary lesions is broad, including infectious, inflammatory, and autoimmune etiologies. Irritant contact dermatitis (ICD) presents similar to ACD, though it is more immediate in onsetand typically demonstrates symptoms of burning and stinging rather than pruritus. Although histopathology is not reliable in differentiating ICD and ACD, it has been shown that focal parakeratosis is associated with ACD, whereas necrotic epidermal keratinocytes are found in ICD.100

Intertrigo presents as large, erythematous, opposing patches or plaques confined to inguinal, submammary, axillary, and/or abdominal folds. Findings of beefy red erythema and peripheral satellite pustules may implicate presence of Candida, which can be identified with potassium hydroxide preparations.

Inverse psoriasis presents as sharply demarcated, erythematous, moist, smooth plaques or patches with minimal scale. The most common area of involvement is the inguinal folds, followed by the axillae, inframammary folds, perianal area, umbilicus, and retroauricular areas. Involvement of the elbows and knees or a positive family history of psoriasis may be useful knowledge in establishing the diagnosis. A biopsy may show dermal eosinophils, epidermal spongiosis, and focal serum in the scale, in addition to features of typical psoriasis plaques.101

Seborrheic dermatitis typically is an erythematous eruption, often with yellowish greasy scale. Simultaneous involvement of the face and scalp may be noted. Although typically a clinical diagnosis, biopsy demonstrates shoulder parakeratosis with follicular plugging and lymphocytic exocytosis.

Hailey-Hailey disease (also called benign familial pemphigus) is an autosomal-dominant genetic condition presenting as moist, malodorous, painful, vegetative plaques, patches, or scaly pustules in flexural areas, frequently with flaccid blisters. Lesions are provoked by traumatic stimuli. Onset occurs in the second to fourth decades and may improve with age. The diagnosis is confirmed by biopsy, which demonstrates acantholysis of the epidermis. The moist superficial patches of Hailey-Hailey disease help distinguish it from comparably drier Darier disease, the other acantholytic disease of the axillae.

Granular parakeratosis (also called hyperkeratotic flexural erythema) is an uncommon dermatosis most often observed in middle-aged women. It presents as red-brown keratotic papules coalescing into plaques, often with overlying scale in intertriginous areas. This disorder may be related to exposure to aluminum, a key component of antiperspirants.102 Diagnosis with a skin biopsy demonstrates granular parakeratosis.

Infections most commonly include erythrasma, tinea, and candidiasis. Erythrasma caused by Corynebacterium minutissimum may present in the axillae and/or groin with sharply demarcated, red-brown patches. Wood lamp examination reveals coral red fluorescence. Tinea corporis, a dermatophyte infection, may present as scaly erythematous plaques with advancing borders and central clearing. Fungal cultures and potassium hydroxide preparations are useful to confirm the diagnosis.

 

 

Pseudofolliculitis barbae most often is thought of as a condition affecting the beard in Black men, but it also may present in individuals of all races who shave the axillary and inguinal regions. Typical features include pruritic inflammatory papules and pustules with surrounding erythema and hyperpigmentation.

Fox-Fordyce disease is a disorder of the apocrine sweat glands that presents as several flesh-colored, perifollicular, monomorphic papules in the axillae. It typically is a disease of young females and also can involve the areola and vulva. Histopathology may show hyperkeratosis, irregular acanthosis, and dilated sweat glands.

Hidradenitis suppurativa is a chronic inflammatory condition that presents with multiple cysts; nodules; abscesses; sinus tract formation; and suppuration of the axillary, anogenital, and sometimes inframammary areas, typically at the onset of puberty. The diagnosis is best supported by history and physical examination, which may be notable for recurrent abscesses, draining tracts, double comedones, and ropelike scarring.

Extramammary Paget disease is a rare malignancy affecting apocrine gland–bearing areas, including axillary and genital regions. It most commonly presents as a unilateral or asymmetric, scaly, erythematous plaque. Histopathology demonstrates Paget cells with abundant clear cytoplasm and pleomorphic nuclei, typically grouped in the lower portion of the epidermis.

Final Thoughts

Axillary dermatoses often can be challenging to diagnose given the range of pathologies that can present in intertriginous areas. Allergic contact dermatitis is a common culprit due to unique anatomical considerations and self-care practices, including shaving/hair removal; use of deodorants, antiperspirants, bodywashes, and clothing; and frictional and moisture influences. The most likely offender among contact allergens is fragrance, but other possibilities to consider include PG, preservatives, aluminum, nickel, and textile dyes. Albeit less common, systemic exposure to allergens may result in SCD and SDRIFE with a rash in intertriginous zones, including the axillae. Additionally, other infectious, inflammatory, and autoimmune etiologies should be considered and ruled out.

Patch testing is the most reliable method to diagnose suspected ACD. Once confirmed, management includes the use of topical steroids and avoidance of the causative agent. Additionally, patients should be informed of the American Contact Dermatitis Society Contact Allergen Management Program (https://www.contactderm.org/patient-support/camp-access), which provides patients with useful information on products that are safe to use based on their patch testing results.

References
  1. Alinaghi F, Bennike NH, Egeberg A, et al. Prevalence of contact allergy in the general population: a systematic review and meta-analysis. Contact Dermatitis. 2019;80:77-85.
  2. Brar KK. A review of contact dermatitis. Ann Allergy Asthma Immunol. 2021;126:32-39.
  3. Evans RL, Marriott RE, Harker M. Axillary skin: biology and care. Int J Cosmet Sci. 2012;34:389-395.
  4. Watkinson A, Lee RS, Moore AE, et al. Is the axilla a distinct skin phenotype? Int J Cosmet Sci. 2007;29:60.
  5. Wu JQ, Kilpatrick-Liverman L. Characterizing the composition of underarm and forearm skin using confocal raman spectroscopy. Int J Cosmet Sci. 2011;33:257-262.
  6. Marti VP, Lee RS, Moore AE, et al. Effect of shaving on axillary stratum corneum. Int J Cosmet Sci. 2003;25:193-198.
  7. Turner GA, Moore AE, Marti VPJ, et al. Impact of shaving and anti-perspirant use on the axillary vault. Int J Cosmet Sci. 2007;29:31-38.
  8. Zhai H, Maibach HI. Skin occlusion and irritant and allergic contact dermatitis: an overview. Contact Dermatitis. 2001;44:201-206.
  9. Lazarov A. Textile dermatitis in patients with contact sensitization in Israel: a 4-year prospective study. J Eur Acad Dermatol Venereol. 2004;18:531-537.
  10. Nelson JL, Mowad CM. Allergic contact dermatitis: patch testing beyond the TRUE Test. J Clin Aesthet Dermatol. 2010;3:36-41.
  11. Zirwas MJ, Moennich J. Antiperspirant and deodorant allergy: diagnosis and management. J Clin Aesthet Dermatol. 2008;1:38-43.
  12. DeKoven JG, Warshaw EM, Reeder MJ, et al. North American Contact Dermatitis Group Patch Test Results: 2019-2020. Dermatitis. 2023;34:90-104.
  13. Eiermann HJ, Larsen W, Maibach HI, et al. Prospective study of cosmetic reactions: 1977-1980. North American Contact Dermatitis Group. J Am Acad Dermatol. 1982;6:909-917.
  14. González-Muñoz P, Conde-Salazar L, Vañó-Galván S. Allergic contact dermatitis caused by cosmetic products. Actas Dermosifiliogr. 2014;105:822-832.
  15. Gerberick GF, Robinson MK, Felter SP, et al. Understanding fragrance allergy using an exposure-based risk assessment approach. Contact Dermatitis. 2001;45:333-340.
  16. Heisterberg MV, Menne T, Andersen KE, et al. Deodorants are the leading cause of allergic contact dermatitis to fragrance ingredients. Contact Dermatitis. 2011;64:258-264.
  17. Johansen JD, Andersen TF, Kjoller M, et al. Identification of risk products for fragrance contact allergy: a case-referent study based on patients’ histories. Am J Contact Dermat. 1998;9:80-86.
  18. Edman B. The influence of shaving method on perfume allergy. Contact Dermatitis. 1994;31:291-292.
  19. Hamza M, Tohid H, Maibach H. Shaving effects on percutaneous penetration: clinical implications. Cutan Ocul Toxicol. 2015;34:335-343.
  20. Geier J, Uter W, Lessmann H, et al. Fragrance mix I and II: results of breakdown tests. Flavour Fragr J. 2015;30:264-274.
  21. Handley J, Burrows D. Allergic contact dermatitis from the synthetic fragrances Lyral and acetyl cedrene in separate underarm deodorant preparations. Contact Dermatitis. 1994;31:288-290.
  22. Hendriks SA, Bousema MT, van Ginkel CJ. Allergic contact dermatitis from the fragrance ingredient Lyral in underarm deodorant. Contact Dermatitis. 1999;41:119.
  23. Jacob SE. Allergic contact dermatitis from lyral in an aerosol deodorant. Dermatitis. 2008;19:216-217.
  24. Gilpin S, Maibach H. Allergic contact dermatitis caused by farnesol: clinical relevance. Cutan Ocul Toxicol. 2010;29:278-287.
  25. Goossens A, Merckx L. Allergic contact dermatitis from farnesol in a deodorant. Contact Dermatitis. 1997;37:179-180.
  26. Schnuch A, Uter W, Geier J, et al. Contact allergy to farnesol in 2021 consecutively patch tested patients. Results of the IVDK. Contact Dermatitis. 2004;50:117-121.
  27. Uter W, Geier J, Schnuch A, et al. Patch test results with patients’ own perfumes, deodorants and shaving lotions: results of the IVDK 1998–2002. J Eur Acad Dermatol Venereol. 2007;21:374-379.
  28. Dittmar D, Schuttelaar MLA. Contact sensitization to hydroperoxides of limonene and linalool: results of consecutive patch testing and clinical relevance. Contact Dermatitis. 2019;80:101-109.
  29. Yazar K, Johnsson S, Lind M-L, et al. Preservatives and fragrances in selected consumer-available cosmetics and detergents. Contact Dermatitis. 2011;64:265-272.
  30. Isaksson M, Karlberg A-T, Nilsson U. Allergic contact dermatitis caused by oxidized linalool in a deodorant. Contact Dermatitis. 2019;81:213-214.
  31. Chen J, Yi Z, Sun R, et al. Analysis of fragrance allergens in personal care products, toys, and water samples: a review. J AOAC Int. 2022;105:396-412.
  32. Larsen WG. Perfume dermatitis. J Am Acad Dermatol. 1985;12:1-9.
  33. Pincelli C, Magni R, Motolese A. Pigmented contact dermatitis from deodorant. Contact Dermatitis. 1993;28:305-306.
  34. Kwong HL, Lim SPR. Pigmented contact dermatitis in the axillae caused by hydroperoxides of limonene. JAAD Case Reports. 2020;6:476-478.
  35. Marks J, Anderson B, DeLeo V. Contact and Occupational Dermatology. 4th ed. Jaypee; 2016.
  36. Johansen JD. Fragrance contact allergy: a clinical review. Am J Clin Dermatol. 2003;4:789-798.
  37. McGowan MA, Scheman A, Jacob SE. Propylene glycol in contact dermatitis: a systematic review. Dermatitis. 2018;29:6-12.
  38. Fiume MM, Bergfeld WF, Belsito DV, et al. Safety assessment of propylene glycol, tripropylene glycol, and PPGs as used in cosmetics. Int J Toxicol. 2012;31(5 suppl):245S-260S.
  39. Farrar CW, Bell HK, King CM. Allergic contact dermatitis from propylene glycol in Efudix cream. Contact Dermatitis. 2003;48:345.
  40. Friedman ES, Friedman PM, Cohen DE, et al. Allergic contact dermatitis to topical minoxidil solution: etiology and treatment. J Am Acad Dermatol. 2002;46:309-312.
  41. Lessmann H, Schnuch A, Geier J, et al. Skin-sensitizing and irritant properties of propylene glycol. Contact Dermatitis. 2005;53:247-259.
  42. Agren-Jonsson S, Magnusson B. Sensitization to propantheline bromide, trichlorocarbanilide and propylene glycol in an antiperspirant. Contact Dermatitis. 1976;2:79-80.
  43. Catanzaro JM, Smith JG Jr. Propylene glycol dermatitis. J Am Acad Dermatol. 1991;24:90-95.
  44. Jacob SE, Scheman A, McGowan MA. Propylene glycol. Dermatitis. 2018;29:3-5.
  45. Carlson S, Gipson K, Nedorost S. Relevance of doubtful (“equivocal”) late patch-test readings. Dermatitis. 2010;21:102-108.
  46. Kullberg SA, Ward JM, Liou YL, et al. Cutaneous reactions to aluminum. Dermatitis. 2020;31:335-349.
  47. Benohanian A. Antiperspirants and deodorants. Clin Dermatol. 2001;19:398-405.
  48. Garg S, Loghdey S, Gawkrodger DJ. Allergic contact dermatitis from aluminum in deodorants. Contact Dermatitis. 2010;62:57-58.
  49. Montemarano AD, Sau P, Johnson FB, et al. Cutaneous granulomas caused by an aluminum-zirconium complex: an ingredient of antiperspirants. J Am Acad Dermatol. 1997;37:496-498.
  50. Rubin L, Slepyan AH, Weber LF, et al. Granulomas of the axillae caused by deodorants. JAMA. 1956;162:953-955.
  51. Williams S, Freemont AJ. Aerosol antiperspirants and axillary granulomata. Br Med J (Clin Res Ed). 1984;288:1651-1652.
  52. Gallego H, Lewis EJ, Crutchfield CE 3rd. Crystal deodorant dermatitis: irritant dermatitis to alum-containing deodorant. Cutis. 1999;64:65-66.
  53. Leventhal JS, Farhadian JA, Miller KE, et al. Crystal deodorant-induced axillary granulomatous dermatitis. Int J Dermatol. 2014;53:e59-e60.
  54. Siemund I, Dahlin J, Hindsén M, et al. Contact allergy to two aluminum salts in consecutively patch-tested dermatitis patients. Dermatitis. 2022;3:31-35.
  55. Bruze M, Netterlid E, Siemund I. Aluminum-allergen of the year 2022. Dermatitis. 2022;33:10-15.
  56. Goiset A, Darrigade A-S, Labrèze C, et al. Aluminum sensitization in a French paediatric patch test population. Contact Dermatitis. 2018;79:382-383.
  57. Admani S, Matiz C, Jacob SE. Nickel allergy—a potential cause of razor dermatitis. Pediatr Dermatol. 2014;31:392-393.
  58. Bibas N, Lassere J, Paul C, et al. Nickel-induced systemic contact dermatitis and intratubal implants: the baboon syndrome revisited. Dermatitis. 2013;24:35-36.
  59. Seidenari S, Manzini BM, Ddanese P. Contact sensitization to textile dyes: description of 100 subjects. Contact Dermatitis. 1991;24:253-258.
  60. Hatch KL, Maibach HI. Textile dye allergic contact dermatitis prevalence. Contact Dermatitis. 2000;42:187-195.
  61. Ryberg K, Isaksson M, Gruvberger B, et al. Contact allergy to textile dyes in southern Sweden. Contact Dermatitis. 2006;54:313-321.
  62. Pratt M, Taraska V. Disperse blue dyes 106 and 124 are common causes of textile dermatitis and should serve as screening allergens for this condition. Dermatitis. 2000;11:30-41.
  63. Seidenari S, Giusti F, Massone F, et al. Sensitization to disperse dyes in a patch test population over a five-year period. Am J Contact Dermat. 2002;13:101-107.
  64. Moreau L, Goossens A. Allergic contact dermatitis associated with reactive dyes in a dark garment: a case report. Contact Dermatitis. 2005;53:150-154.
  65. Svedman C, Engfeldt M, Malinauskiene L. Textile contact dermatitis: how fabrics can induce dermatitis. Curr Treat Options Allergy. 2019;6:103-111.
  66. Jacob SE, Zapolanski T. Systemic contact dermatitis. Dermatitis. 2008;19:9-15.
  67. Hindsén M, Bruze M, Christensen OB. Flare-up reactions after oral challenge with nickel in relation to challenge dose and intensity and time of previous patch test reactions. J Am Acad Dermatol. 2001;44:616-623.
  68. Winnicki M, Shear NH. A systematic approach to systemic contact dermatitis and symmetric drug-related intertriginous and flexural exanthema (SDRIFE): a closer look at these conditions and an approach to intertriginous eruptions. Am J Clin Dermatol. 2011;12:171-180.
  69. Kalita BJ, Das S, Dutta B. Itraconazole-induced symmetrical drug-related intertriginous and flexural exanthema (SDRIFE): a rare occurrence. Int J Dermatol. 2020;59:e419-e421.
  70. Salam TN, Fowler JF Jr. Balsam-related systemic contact dermatitis. J Am Acad Dermatol. 2001;45:377-381.
  71. Ramachandran V, Cline A, Summey B, et al. Systemic contact dermatitis related to alcoholic beverage consumption. Dermatol Online J. 2019;25:13030/qt3zg853qv.
  72. Moreno-Ramírez D, García-Bravo B, Pichardo AR, et al. Baboon syndrome in childhood: easy to avoid, easy to diagnose, but the problem continues. Pediatr Dermatol. 2004;21:250-253.
  73. Dou X, Liu L-L, Zhu X-J. Nickel-elicited systemic contact dermatitis. Contact Dermatitis. 2003;48:126-129.
  74. Möller H, Ohlsson K, Linder C, et al. The flare-up reactions after systemic provocation in contact allergy to nickel and gold. Contact Dermatitis. 1999;40:200-204.
  75. Andersen KE, Hjorth N, Menné T. The baboon syndrome: systemically-induced allergic contact dermatitis. Contact Dermatitis. 1984;10:97-100.
  76. Häusermann P, Harr T, Bircher AJ. Baboon syndrome resulting from systemic drugs: is there strife between SDRIFE and allergic contact dermatitis syndrome? Contact Dermatitis. 2004;51:297-310.
  77. Tan MG, Pratt MD, Burns BF, et al. Baboon syndrome from mercury showing leukocytoclastic vasculitis on biopsy. Contact Dermatitis. 2020;83:415-417.
  78. Handisurya A, Stingl G, Wöhrl S. SDRIFE (baboon syndrome) induced by penicillin. Clin Exp Dermatol. 2009;34:355-357.
  79. Akay BN, Sanli H. Symmetrical drug-related intertriginous and flexural exanthem due to oral risperidone. Pediatr Dermatol. 2009;26:214-216.
  80. Amaro C, Santos R, Cardoso J. Contact allergy to methylisothiazolinone in a deodorant. Contact Dermatitis. 2011;64:298-299.
  81. Goh CL. Dermatitis from chlorphenesin in a deodorant. Contact Dermatitis. 1987;16:287.
  82. Taghipour K, Tatnall F, Orton D. Allergic axillary dermatitis due to hydrogenated castor oil in a deodorant. Contact Dermatitis. 2008;58:168-169.
  83. Sheu M, Simpson EL, Law S V, et al. Allergic contact dermatitis from a natural deodorant: a report of 4 cases associated with lichen acid mix allergy. J Am Acad Dermatol. 2006;55:332-337.
  84. Pastor-Nieto M-A, Gatica-Ortega M-E, Alcántara-Nicolás F-D-A, et al. Allergic contact dermatitis resulting from cetyl PEG/PPG-10/1 dimethicone in a deodorant cream. Contact Dermatitis. 2018;78:236-239.
  85. Corazza M, Lombardi AR, Virgili A. Non-eczematous urticarioid allergic contact dermatitis due to Eumulgin L in a deodorant. Contact Dermatitis. 1997;36:159-160.
  86. van Ketel WG. Allergic contact dermatitis from propellants in deodorant sprays in combination with allergy to ethyl chloride. Contact Dermatitis. 1976;2:115-119.
  87. Shmunes E, Levy EJ. Quaternary ammonium compound contact dermatitis from a deodorant. Arch Dermatol. 1972;105:91-93.
  88. Bruze M, Johansen JD, Andersen KE, et al. Deodorants: an experimental provocation study with cinnamic aldehyde. J Am Acad Dermatol. 2003;48:194-200.
  89. Hann S, Hughes TM, Stone NM. Flexural allergic contact dermatitis to benzalkonium chloride in antiseptic bath oil. Br J Dermatol. 2007;157:795-798.
  90. Aeling JL, Panagotacos PJ, Andreozzi RJ. Allergic contact dermatitis to vitamin E aerosol deodorant. Arch Dermatol. 1973;108:579-580.
  91. Cotton CH, Duah CG, Matiz C. Allergic contact dermatitis due to methylisothiazolinone in a young girl’s laundry detergent. Pediatr Dermatol. 2017;34:486-487.
  92. Magnano M, Silvani S, Vincenzi C, et al. Contact allergens and irritants in household washing and cleaning products. Contact Dermatitis. 2009;61:337-341.
  93. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. Contact Dermatitis. 2020;82:313-314.
  94. Iammatteo M, Akenroye A, Jariwala S, et al. Severe contact dermatitis due to ethylenediamine dihydrochloride in nystatin cream. J Allergy Clin Immunol Pract. 2017;5:1448-1450.
  95. Coskey RJ, Bryan HG. Contact dermatitis due to methylprednisolone. JAMA. 1967;199:136.
  96. Peterson MY, Han J, Warshaw EM. Allergic contact dermatitis from dipropylene glycol in hydrocortisone lotion. Contact Dermatitis. 2022;87:112-114.
  97. Ferreira O, Cruz MJ, Mota A, et al. Erythema multiforme-like lesions revealing allergic contact dermatitis to exotic woods. Cutan Ocul Toxicol. 2012;31:61-63.
  98. Abraham NF, Feldman SR, Vallejos Q, et al. Contact dermatitis in tobacco farmworkers. Contact Dermatitis. 2007;57:40-43.
  99. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. J Am Acad Dermatol. 2016;74:1043-1054.
  100. Frings VG, Böer-Auer A, Breuer K. Histomorphology and immunophenotype of eczematous skin lesions revisited-skinbiopsies are not reliable in differentiating allergic contact dermatitis, irritant contact dermatitis, and atopic dermatitis. Am J Dermatopathol. 2018;40:7-16.
  101. Knabel M, Mudaliar K. Histopathologic features of inverse psoriasis. J Cutan Pathol. 2022;49:246-251.
  102. Fujii M, Kishibe M, Honma M, et al. Aluminum chloride-induced apoptosis leads to keratinization arrest and granular parakeratosis. Am J Dermatopathol. 2020;42:756-761.
Article PDF
Author and Disclosure Information

Dr. Musicante is from The University of Tennessee Health Science Center College of Medicine, Memphis. Drs. Cohen and Milam are from the Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York.

Drs. Musicante and Milam report no conflict of interest. Dr. Cohen has been a consultant for and received honoraria from Cosmetic Ingredient Review; Ferndale Laboratories, Inc; FIDE; LEO Pharma; Medimetriks; Novartis (past); SFJ Pharmaceuticals, Inc (past); and UCB. He also owns stock or has stock options in Evommune, Kadmon (past), and Timber Pharmaceuticals, and is on the board of directors for Evommune, Kadmon (past), and Timber Pharmaceuticals.

Correspondence: Emily C. Milam, MD, 240 E 38th St, Floor 12, New York, NY 10016 (Emily.Milam@nyulangone.org).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
35-42
Sections
Author and Disclosure Information

Dr. Musicante is from The University of Tennessee Health Science Center College of Medicine, Memphis. Drs. Cohen and Milam are from the Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York.

Drs. Musicante and Milam report no conflict of interest. Dr. Cohen has been a consultant for and received honoraria from Cosmetic Ingredient Review; Ferndale Laboratories, Inc; FIDE; LEO Pharma; Medimetriks; Novartis (past); SFJ Pharmaceuticals, Inc (past); and UCB. He also owns stock or has stock options in Evommune, Kadmon (past), and Timber Pharmaceuticals, and is on the board of directors for Evommune, Kadmon (past), and Timber Pharmaceuticals.

Correspondence: Emily C. Milam, MD, 240 E 38th St, Floor 12, New York, NY 10016 (Emily.Milam@nyulangone.org).

Author and Disclosure Information

Dr. Musicante is from The University of Tennessee Health Science Center College of Medicine, Memphis. Drs. Cohen and Milam are from the Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York.

Drs. Musicante and Milam report no conflict of interest. Dr. Cohen has been a consultant for and received honoraria from Cosmetic Ingredient Review; Ferndale Laboratories, Inc; FIDE; LEO Pharma; Medimetriks; Novartis (past); SFJ Pharmaceuticals, Inc (past); and UCB. He also owns stock or has stock options in Evommune, Kadmon (past), and Timber Pharmaceuticals, and is on the board of directors for Evommune, Kadmon (past), and Timber Pharmaceuticals.

Correspondence: Emily C. Milam, MD, 240 E 38th St, Floor 12, New York, NY 10016 (Emily.Milam@nyulangone.org).

Article PDF
Article PDF

Approximately 20% of the general population has a contact allergy.1 Allergic contact dermatitis (ACD) is a delayed type IV hypersensitivity reaction mediated by T lymphocytes.2 Axillary ACD presentation is variable but typically includes an eczematous eruption with erythematous scaly patches or plaques. Common products in contact with the axillae include deodorants, antiperspirants, razors, bodywash, and clothing.

Axillary skin is distinct from skin elsewhere on the body due to both anatomical characteristics and unique human self-care practices. Axillary skin has reduced barrier function, faster stratum corneum turnover, and altered lipid levels.3-5 Moreover, the axillae often are subject to shaving or other hair removal practices that alter the local environment, as layers of stratum corneum and hair are mechanically removed, which causes irritation and predisposes the skin to enhanced sensitivity to topical exposures.6,7 With the abundance of apocrine and eccrine glands, the axillae are prone to sweat, which also can accentuate contact allergy.2,3 Other factors, such as occlusion and friction, contribute to axillary contact allergy.8,9

Patch testing is the gold standard for the diagnosis of ACD and aids in identification of culprit allergens. A thorough patient history and examination of the rash distribution may provide further clues; for example, dermatitis due to a deodorant typically affects the vault, whereas textile dye dermatitis tends to spare the vault.10,11 Baseline-limited patch testing detects up to two-thirds of clinically relevant allergens.12 Therefore, patients may require subsequent testing with supplemental allergens.

The differential diagnosis for axillary lesions is broad—including inflammatory diseases such as irritant contact dermatitis and hidradenitis suppurativa, genetic disorders such as Hailey-Hailey disease, and infectious causes such as erythrasma—but may be narrowed with a thorough physical examination and patient history, histopathology, bedside diagnostic techniques (eg, scrapings and Wood lamp examination), and patch testing. Systemic contact dermatitis (SCD) or symmetrical drug-related intertriginous and flexural exanthema (SDRIFE) also may be suspected in cases of intertriginous dermatoses.

We review the potential allergens in products used on the axillae as well as the management of axillary ACD. We also discuss axillary dermatitis as a manifestation of SCD and SDRIFE.

Top Allergens in Products Used on the Axillae

Fragrance—A 1982 North American Contact Dermatitis Group study on cosmetic products identified fragrances as the most common cause of ACD,13 and this trend continues to hold true with more recent data.14 The incidence of fragrance allergy may be increasing, with positive patch tests to a fragrance chemical in 10% of patch test clinic populations.15 Fragrances are a ubiquitous ingredient in deodorants and antiperspirants, which are important sources implicated in the development and elicitation of fragrance ACD.16 One study found that fragrance was present in 97 of 107 (90%) deodorants available at Walgreens pharmacies.11

In a study of patients with a history of an axillary rash caused by a deodorant spray, Johansen et al17 reported that the likelihood of fragrance allergy is increased by a factor of 2.4. This risk of developing a fragrance allergy may be exacerbated in those who shave; Edman18 reported that the odds ratio of developing a fragrance allergy among men who shave their beards was 2.9. Although there are no specific data on the effects of shaving on ACD, shaving in general can induce localized irritation and increase percutaneous absorption.19

 

 

The individual fragrance components in deodorants most likely to cause ACD include hydroxycitronellal, eugenol, and geraniol—all constituent ingredients in patch test formulations of fragrance mixture I.11,20 Other common fragrance allergens associated with ACD include hydroxymethylpentylcyclohexenecarboxaldehyde, farnesol, and balsam of Peru.21-27 Hydroperoxides of limonene and linalool, common fragrances in detergents and personal care products, are increasingly recognized as contact allergens and have been reported to cause axillary ACD from deodorants.28-30

Dermatitis involving the bilateral axillary vaults wherever deodorant or antiperspirant was directly applied is the most common presentation of ACD due to fragrance (Figure 1).17 An eczematous eruption is common, though scale may be less apparent than in nonflexural regions. Axillary ACD secondary to fragrances also may result from use of fragranced laundry detergents, fabric softeners, soaps, and perfumes, and may spare the vaults.10,29,31,32 Less common presentations of axillary ACD due to fragrance include pigmented dermatoses; for example, ACD from an antiperspirant containing hydroperoxide of limonene presented as hyperpigmented patches with minimal erythema and scaling in the edges of the axillary folds.33,34

Musicante_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Allergic%20contact%20dermatitis%20of%20the%20axillary%20vault%20secondary%20to%20use%20of%20scented%20antiperspirant%2Fdeodorant%20in%20a%20patient%20with%20positive%20patch%20test%20results%20to%20propylene%20glycol%2C%20balsam%20of%20Peru%2C%20and%20quaternium-15.%3C%2Fp%3E

Diagnosis of a fragrance ACD typically is made with a standard patch test series including fragrance mixture I and balsam of Peru, which may detect 75% and 50% of fragrance sensitivities, respectively.35 Patch testing may be followed with a repeated open application test of the product in question.36 Additionally, it may be appropriate to test for other fragrance allergens including balsam of Tolu, fragrance mixture II, lichen acid mix, and hydroxyperoxides of linalool and limonene (among other botanicals) if standard patch testing is negative and suspicion of fragrance ACD remains elevated.11

Propylene Glycol—Propylene glycol (PG)—a versatile substance that functions as a solvent, humectant, emulsifier, stabilizer, and antimicrobial—is the second most common contact allergen present in deodorants.11 It is prevalent in both personal care and household products, including deodorants, cosmetics, foods, toothpaste, cleaning agents, and detergents.11,37 Propylene glycol is both an allergen and an irritant. Among deodorants/antiperspirants, PG is both a common irritant and allergen, as its concentration may be particularly high (as much as 73%).38 One commonly reported example of PG contact dermatitis is from the topical medicament minoxidil.39,40

Patch testing data have demonstrated a positivity rate for PG ranging between 0.1% to 3.8%. The variability in these findings likely is due to differences in the tested concentrations of PG, as higher concentrations sometimes required to elicit an allergic reaction also may create a stronger irritation effect.41 Propylene glycol irritancy and the occlusive nature of the axillae may enhance sensitization to other allergens, as demonstrated by Agren-Jonsson and Magnusson,42 who reported sensitization to propantheline bromide and trichlorocarbanilide in patients who used a lotion with 90% PG. Many PG-containing products beyond deodorants/antiperspirants may be applied to the axillae, including steroid creams, lotions, shaving creams, and bodywashes.38,43

The diagnosis of PG allergy via patch testing is challenging and at times controversial given its irritant nature. False-positive irritant reactions have been documented, characterized by a weak reaction at 48 hours that is absent by 96 hours (decrescendo reaction). A reaction may not appear until 96 hours (crescendo reaction), which typically indicates a true contact allergy but in the case of PG also may be the substance acting as a “late irritant.”44 Fast (<24 hours) and well-demarcated reactions suggest irritation.45 Regardless, reactions to PG on patch testing, even those regarded as weak, may be considered relevant in consideration of the clinical context.37

Aluminum—Aluminum is the active ingredient in most antiperspirants, typically in the form of aluminum chloride, aluminum chlorohydrate, aluminum zirconium trichlorohydrex gly, or aluminum zirconium tetrachlorohydrex gly.46 Aluminum mechanically obstructs the eccrine glands to reduce sweat.47 Although aluminum is an uncommon allergen, a possible presentation of aluminum allergy is axillary vault dermatitis secondary to antiperspirant use.46 Another potential manifestation is a ringlike reaction to the Finn Chambers (SmartPractice) used in patch testing.46 In one case of aluminum-induced axillary dermatitis, a 28-year-old woman presented with eczema of the axillae, and subsequent patch testing revealed an allergy to aluminum chloride. The rash resolved upon cessation of use of an aluminum-containing deodorant.48

 

 

Aluminum has been reported to cause granulomatous dermatitis in the axillae. This reaction typically presents as red-brown, pruritic papules limited to the area in which deodorant was applied, with histopathology revealing epithelioid granulomas.49-51

Alum deodorants—considered a natural alternative—contain aluminum bound to potassium or ammonium in the form of a crystal or powder. Alum crystal deodorants have been reported to cause both a typical erythematous pruritic dermatitis as well as a granulomatous dermatitis with red-brown papules.52,53 The granulomatous dermatitis caused by either form of aluminum resolves with avoidance and use of topical steroids or topical tacrolimus.49,50,52,53

The diagnosis of aluminum ACD via patch testing may be identified with empty Finn Chambers, which are metallic aluminum, or with patch placement of aluminum chloride hexahydrate, though the former is only positive in patients with a strong allergy.54,55 In 2022, aluminum was named Allergen of the Year by the American Contact Dermatitis Society, with recommendations to conduct patch testing with aluminum chloride hexahydrate 10% rather than the traditional 2% to increase diagnostic yield.55 Additionally, it is recommended that aluminum be included in baseline patch testing for children due to the high prevalence of aluminum allergy in children and early exposure via childhood vaccines.54-56 In patients with aluminum allergy, providers may suggest purchasing aluminum-free deodorants or provide recipes for homemade deodorant that includes ingredients such as arrowroot powder, cornstarch, and diatomaceous earth.46

Nickel—Nickel is the most commonly identified contact allergen on patch testing yet an infrequent cause of axillary dermatitis. A case report from 2014 described axillary dermatitis in a woman that worsened during a positive patch test to nickel. Improvement was noted when the patient switched to titanium shaving razors.57 Nickel allergy also may present in the form of SCD. In one report, a woman developed dermatitis of the flexural areas, including the axillae, 3 months after undergoing a sterilization procedure in which nickel-containing tubal implants were placed.58 Patch testing revealed a positive reaction to nickel. The patient experienced complete resolution of the steroid-resistant dermatitis following removal of the implants via salpingectomy.58

Musicante_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Textile%20allergic%20contact%20dermatitis%20secondary%20to%20a%20deeply%20dyed%20blue%20sweater%20in%20a%20patient%20with%20positive%20patch%20test%20results%20to%20disperse%20blue%20106%2C%20disperse%20blue%20124%2C%20textile%20dye%20mix%2C%20formaldehyde%2C%20and%20methyldibromo%20glutaronitrile%2C%20among%20other%20allergens.%20The%20dermatitis%20involved%20the%20bilateral%20axillary%20rim%20and%20spared%20the%20vault.%3C%2Fp%3E

Textile Dye—In contrast to dermatitis caused by deodorants/antiperspirants, contact allergy to textile dyes presents as dermatitis involving the axillary borders but sparing the axillary vaults (Figures 2 and 3).10 Other potential presentations of textile dye dermatitis include erythema multiforme–like eruptions and erythematous wheal–type reactions.59 Textile dyes are classified as disperse vs nondisperse, with the majority of contact dermatoses caused by disperse dyes, specifically Disperse Orange 1, blue 106, and blue 124.60-62 Ryberg et al61 found that the axilla is one of the more common locations to be affected by textile dye allergy, particularly in women, which was further supported by Seidenari et al,63 who found that skin folds were affected in 27% of study participants allergic to textile dyes (N=437), a finding that is likely due to friction, sweat, and occlusion.62 In one case report of a patient with dermatitis caused by reactive dyes, the garment required 3 washes before the patient experienced resolution of dermatitis.64 For patients with textile dye dermatitis, mitigation strategies include washing clothing before wearing, especially for darkly dyed items; avoiding tight clothing; wearing garments made of cotton, wool, silk, or linen; and choosing light-colored garments.9,64,65

Musicante_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Allergic%20contact%20dermatitis%20of%20the%20axilla%20suspected%20to%20be%20secondary%20to%20black%20textile%20dyes.%20The%20dermatitis%20resolved%20completely%20with%20avoidance%20of%20tightly%20fitted%20black%20clothing.%3C%2Fp%3E

Axillary Dermatitis as a Manifestation of SCD and SDRIFE

Systemic contact dermatitis occurs when an individual who was previously sensitized to a particular allergen develops ACD of the skin with systemic exposure to that allergen or immunochemically related allergens. Exposure may occur via ingestion, inhalation, intravenous, intramuscular, and transepidermal routes.66 Systemic contact dermatitis manifests in a variety of ways, including focal flares at sites of prior contact dermatitis (recall reaction), vesicular hand dermatitis, intertriginous eruptions including axillary dermatitis, and generalized eruptions.67

Systemic contact dermatitis rarely involves systemic symptoms, and onset typically is within days of exposure. The 3 most common groups of allergens causing SCD are metals, medications, and plants and herbals.68 These allergens have all been reported to cause axillary dermatitis via SCD.58,69,70 Foods containing balsam of Peru that may lead to SCD include citrus, chocolate, tomato, and certain alcohols.70,71 Patients with a positive patch test to balsam of Peru may experience improvement of their dermatitis after reduction of balsam of Peru–rich foods from their diet.70 Metals implicated in SCD include mercury, nickel, and gold.72-74 Finally, PG ingestion also has been implicated in cases of SCD.37

 

 

Symmetrical drug-related intertriginous and flexural exanthema is another condition that presents as intertriginous dermatitis and differs from SCD in that the eruption does not require presensitization; there may be no known prior exposure to the agent causing dermatitis. Historically, SDRIFE was described as baboon syndrome because of its frequent involvement of the buttocks with diffuse, well-demarcated, erythematous dermatitis resembling that of a baboon. This term is no longer used due to its insensitive nature and incomplete depiction of SDRIFE, which can affect body sites other than the buttocks.68,75,76 Specific criteria to make this diagnosis include sharply demarcated and/or V-shaped erythema of the gluteal/perianal area, involvement of at least 1 other intertriginous or flexural region, symmetry of affected areas, and an absence of systemic symptoms.76 There also may be papules, pustules, and vesicles present in affected areas. Symmetrical drug-related intertriginous and flexural exanthema most often is caused by β-lactam antibiotics, but other associated drugs include chemotherapeutic agents, such as mitomycin C.76

Histopathology of both SCD and SDRIFE is variable and typically nonspecific, often revealing epidermal spongiosis and a perivascular mononuclear cell infiltrate with occasional neutrophils and eosinophils.76 A case of SCD to mercury presenting as intertriginous dermatitis demonstrated a leukocytoclastic vasculitis pattern on biopsy.77

Systemic contact dermatitis is diagnosed via a patch test, while SDRIFE typically has a negative patch test result and requires oral rechallenge testing, which reproduces the rash within hours.78,79

Additional Allergens Causing Axillary ACD

Although fragrance is the most common allergen in deodorants, other ingredients have been shown to cause axillary ACD (Table).80-90 In addition to these ingredients, allergens not previously mentioned that may be present in deodorants include lanolin, essential oils, and parabens.11 Methylisothiazolinone in laundry detergent also has been found to instigate ACD.91 Fragrances and preservatives in laundry detergents also may contribute to dermatitis.92

CT113001035_Table.jpg

Other products that have caused axillary contact dermatitis include topical exposure to medicaments including clindamycin,93 ethylenediamine in nystatin cream,94 methylprednisolone acetate95 and dipropylene glycol in a hydrocortisone lotion,96 wood dusts from tropical hardwoods,97 and tobacco.98

Management of ACD

The most effective strategy in the management of patients with contact dermatitis is avoidance of the offending agent. Additionally, clinicians may recommend the use of topical steroids and/or calcineurin inhibitors to hasten resolution.2

For patients with contact dermatitis, a clinician may recommend product substitutions with few potential allergens to use prior to patch testing. Patients with a fragrance allergy should look for products specifically labeled as “fragrance free” rather than “hypoallergenic” or “unscented,” as the latter two may still contain minimal amounts of fragrance.35 Patients should be educated on the functions of the allergens to which they are allergic so they may adequately avoid potential sources of contact.99 For suspected textile dye dermatitis, instructing patients to wash clothing before wearing and to avoid synthetic fabrics, dark dyes, and tightly fitted clothing may help.9,64,65

 

 

Differential Diagnosis

The differential diagnosis for axillary lesions is broad, including infectious, inflammatory, and autoimmune etiologies. Irritant contact dermatitis (ICD) presents similar to ACD, though it is more immediate in onsetand typically demonstrates symptoms of burning and stinging rather than pruritus. Although histopathology is not reliable in differentiating ICD and ACD, it has been shown that focal parakeratosis is associated with ACD, whereas necrotic epidermal keratinocytes are found in ICD.100

Intertrigo presents as large, erythematous, opposing patches or plaques confined to inguinal, submammary, axillary, and/or abdominal folds. Findings of beefy red erythema and peripheral satellite pustules may implicate presence of Candida, which can be identified with potassium hydroxide preparations.

Inverse psoriasis presents as sharply demarcated, erythematous, moist, smooth plaques or patches with minimal scale. The most common area of involvement is the inguinal folds, followed by the axillae, inframammary folds, perianal area, umbilicus, and retroauricular areas. Involvement of the elbows and knees or a positive family history of psoriasis may be useful knowledge in establishing the diagnosis. A biopsy may show dermal eosinophils, epidermal spongiosis, and focal serum in the scale, in addition to features of typical psoriasis plaques.101

Seborrheic dermatitis typically is an erythematous eruption, often with yellowish greasy scale. Simultaneous involvement of the face and scalp may be noted. Although typically a clinical diagnosis, biopsy demonstrates shoulder parakeratosis with follicular plugging and lymphocytic exocytosis.

Hailey-Hailey disease (also called benign familial pemphigus) is an autosomal-dominant genetic condition presenting as moist, malodorous, painful, vegetative plaques, patches, or scaly pustules in flexural areas, frequently with flaccid blisters. Lesions are provoked by traumatic stimuli. Onset occurs in the second to fourth decades and may improve with age. The diagnosis is confirmed by biopsy, which demonstrates acantholysis of the epidermis. The moist superficial patches of Hailey-Hailey disease help distinguish it from comparably drier Darier disease, the other acantholytic disease of the axillae.

Granular parakeratosis (also called hyperkeratotic flexural erythema) is an uncommon dermatosis most often observed in middle-aged women. It presents as red-brown keratotic papules coalescing into plaques, often with overlying scale in intertriginous areas. This disorder may be related to exposure to aluminum, a key component of antiperspirants.102 Diagnosis with a skin biopsy demonstrates granular parakeratosis.

Infections most commonly include erythrasma, tinea, and candidiasis. Erythrasma caused by Corynebacterium minutissimum may present in the axillae and/or groin with sharply demarcated, red-brown patches. Wood lamp examination reveals coral red fluorescence. Tinea corporis, a dermatophyte infection, may present as scaly erythematous plaques with advancing borders and central clearing. Fungal cultures and potassium hydroxide preparations are useful to confirm the diagnosis.

 

 

Pseudofolliculitis barbae most often is thought of as a condition affecting the beard in Black men, but it also may present in individuals of all races who shave the axillary and inguinal regions. Typical features include pruritic inflammatory papules and pustules with surrounding erythema and hyperpigmentation.

Fox-Fordyce disease is a disorder of the apocrine sweat glands that presents as several flesh-colored, perifollicular, monomorphic papules in the axillae. It typically is a disease of young females and also can involve the areola and vulva. Histopathology may show hyperkeratosis, irregular acanthosis, and dilated sweat glands.

Hidradenitis suppurativa is a chronic inflammatory condition that presents with multiple cysts; nodules; abscesses; sinus tract formation; and suppuration of the axillary, anogenital, and sometimes inframammary areas, typically at the onset of puberty. The diagnosis is best supported by history and physical examination, which may be notable for recurrent abscesses, draining tracts, double comedones, and ropelike scarring.

Extramammary Paget disease is a rare malignancy affecting apocrine gland–bearing areas, including axillary and genital regions. It most commonly presents as a unilateral or asymmetric, scaly, erythematous plaque. Histopathology demonstrates Paget cells with abundant clear cytoplasm and pleomorphic nuclei, typically grouped in the lower portion of the epidermis.

Final Thoughts

Axillary dermatoses often can be challenging to diagnose given the range of pathologies that can present in intertriginous areas. Allergic contact dermatitis is a common culprit due to unique anatomical considerations and self-care practices, including shaving/hair removal; use of deodorants, antiperspirants, bodywashes, and clothing; and frictional and moisture influences. The most likely offender among contact allergens is fragrance, but other possibilities to consider include PG, preservatives, aluminum, nickel, and textile dyes. Albeit less common, systemic exposure to allergens may result in SCD and SDRIFE with a rash in intertriginous zones, including the axillae. Additionally, other infectious, inflammatory, and autoimmune etiologies should be considered and ruled out.

Patch testing is the most reliable method to diagnose suspected ACD. Once confirmed, management includes the use of topical steroids and avoidance of the causative agent. Additionally, patients should be informed of the American Contact Dermatitis Society Contact Allergen Management Program (https://www.contactderm.org/patient-support/camp-access), which provides patients with useful information on products that are safe to use based on their patch testing results.

Approximately 20% of the general population has a contact allergy.1 Allergic contact dermatitis (ACD) is a delayed type IV hypersensitivity reaction mediated by T lymphocytes.2 Axillary ACD presentation is variable but typically includes an eczematous eruption with erythematous scaly patches or plaques. Common products in contact with the axillae include deodorants, antiperspirants, razors, bodywash, and clothing.

Axillary skin is distinct from skin elsewhere on the body due to both anatomical characteristics and unique human self-care practices. Axillary skin has reduced barrier function, faster stratum corneum turnover, and altered lipid levels.3-5 Moreover, the axillae often are subject to shaving or other hair removal practices that alter the local environment, as layers of stratum corneum and hair are mechanically removed, which causes irritation and predisposes the skin to enhanced sensitivity to topical exposures.6,7 With the abundance of apocrine and eccrine glands, the axillae are prone to sweat, which also can accentuate contact allergy.2,3 Other factors, such as occlusion and friction, contribute to axillary contact allergy.8,9

Patch testing is the gold standard for the diagnosis of ACD and aids in identification of culprit allergens. A thorough patient history and examination of the rash distribution may provide further clues; for example, dermatitis due to a deodorant typically affects the vault, whereas textile dye dermatitis tends to spare the vault.10,11 Baseline-limited patch testing detects up to two-thirds of clinically relevant allergens.12 Therefore, patients may require subsequent testing with supplemental allergens.

The differential diagnosis for axillary lesions is broad—including inflammatory diseases such as irritant contact dermatitis and hidradenitis suppurativa, genetic disorders such as Hailey-Hailey disease, and infectious causes such as erythrasma—but may be narrowed with a thorough physical examination and patient history, histopathology, bedside diagnostic techniques (eg, scrapings and Wood lamp examination), and patch testing. Systemic contact dermatitis (SCD) or symmetrical drug-related intertriginous and flexural exanthema (SDRIFE) also may be suspected in cases of intertriginous dermatoses.

We review the potential allergens in products used on the axillae as well as the management of axillary ACD. We also discuss axillary dermatitis as a manifestation of SCD and SDRIFE.

Top Allergens in Products Used on the Axillae

Fragrance—A 1982 North American Contact Dermatitis Group study on cosmetic products identified fragrances as the most common cause of ACD,13 and this trend continues to hold true with more recent data.14 The incidence of fragrance allergy may be increasing, with positive patch tests to a fragrance chemical in 10% of patch test clinic populations.15 Fragrances are a ubiquitous ingredient in deodorants and antiperspirants, which are important sources implicated in the development and elicitation of fragrance ACD.16 One study found that fragrance was present in 97 of 107 (90%) deodorants available at Walgreens pharmacies.11

In a study of patients with a history of an axillary rash caused by a deodorant spray, Johansen et al17 reported that the likelihood of fragrance allergy is increased by a factor of 2.4. This risk of developing a fragrance allergy may be exacerbated in those who shave; Edman18 reported that the odds ratio of developing a fragrance allergy among men who shave their beards was 2.9. Although there are no specific data on the effects of shaving on ACD, shaving in general can induce localized irritation and increase percutaneous absorption.19

 

 

The individual fragrance components in deodorants most likely to cause ACD include hydroxycitronellal, eugenol, and geraniol—all constituent ingredients in patch test formulations of fragrance mixture I.11,20 Other common fragrance allergens associated with ACD include hydroxymethylpentylcyclohexenecarboxaldehyde, farnesol, and balsam of Peru.21-27 Hydroperoxides of limonene and linalool, common fragrances in detergents and personal care products, are increasingly recognized as contact allergens and have been reported to cause axillary ACD from deodorants.28-30

Dermatitis involving the bilateral axillary vaults wherever deodorant or antiperspirant was directly applied is the most common presentation of ACD due to fragrance (Figure 1).17 An eczematous eruption is common, though scale may be less apparent than in nonflexural regions. Axillary ACD secondary to fragrances also may result from use of fragranced laundry detergents, fabric softeners, soaps, and perfumes, and may spare the vaults.10,29,31,32 Less common presentations of axillary ACD due to fragrance include pigmented dermatoses; for example, ACD from an antiperspirant containing hydroperoxide of limonene presented as hyperpigmented patches with minimal erythema and scaling in the edges of the axillary folds.33,34

Musicante_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Allergic%20contact%20dermatitis%20of%20the%20axillary%20vault%20secondary%20to%20use%20of%20scented%20antiperspirant%2Fdeodorant%20in%20a%20patient%20with%20positive%20patch%20test%20results%20to%20propylene%20glycol%2C%20balsam%20of%20Peru%2C%20and%20quaternium-15.%3C%2Fp%3E

Diagnosis of a fragrance ACD typically is made with a standard patch test series including fragrance mixture I and balsam of Peru, which may detect 75% and 50% of fragrance sensitivities, respectively.35 Patch testing may be followed with a repeated open application test of the product in question.36 Additionally, it may be appropriate to test for other fragrance allergens including balsam of Tolu, fragrance mixture II, lichen acid mix, and hydroxyperoxides of linalool and limonene (among other botanicals) if standard patch testing is negative and suspicion of fragrance ACD remains elevated.11

Propylene Glycol—Propylene glycol (PG)—a versatile substance that functions as a solvent, humectant, emulsifier, stabilizer, and antimicrobial—is the second most common contact allergen present in deodorants.11 It is prevalent in both personal care and household products, including deodorants, cosmetics, foods, toothpaste, cleaning agents, and detergents.11,37 Propylene glycol is both an allergen and an irritant. Among deodorants/antiperspirants, PG is both a common irritant and allergen, as its concentration may be particularly high (as much as 73%).38 One commonly reported example of PG contact dermatitis is from the topical medicament minoxidil.39,40

Patch testing data have demonstrated a positivity rate for PG ranging between 0.1% to 3.8%. The variability in these findings likely is due to differences in the tested concentrations of PG, as higher concentrations sometimes required to elicit an allergic reaction also may create a stronger irritation effect.41 Propylene glycol irritancy and the occlusive nature of the axillae may enhance sensitization to other allergens, as demonstrated by Agren-Jonsson and Magnusson,42 who reported sensitization to propantheline bromide and trichlorocarbanilide in patients who used a lotion with 90% PG. Many PG-containing products beyond deodorants/antiperspirants may be applied to the axillae, including steroid creams, lotions, shaving creams, and bodywashes.38,43

The diagnosis of PG allergy via patch testing is challenging and at times controversial given its irritant nature. False-positive irritant reactions have been documented, characterized by a weak reaction at 48 hours that is absent by 96 hours (decrescendo reaction). A reaction may not appear until 96 hours (crescendo reaction), which typically indicates a true contact allergy but in the case of PG also may be the substance acting as a “late irritant.”44 Fast (<24 hours) and well-demarcated reactions suggest irritation.45 Regardless, reactions to PG on patch testing, even those regarded as weak, may be considered relevant in consideration of the clinical context.37

Aluminum—Aluminum is the active ingredient in most antiperspirants, typically in the form of aluminum chloride, aluminum chlorohydrate, aluminum zirconium trichlorohydrex gly, or aluminum zirconium tetrachlorohydrex gly.46 Aluminum mechanically obstructs the eccrine glands to reduce sweat.47 Although aluminum is an uncommon allergen, a possible presentation of aluminum allergy is axillary vault dermatitis secondary to antiperspirant use.46 Another potential manifestation is a ringlike reaction to the Finn Chambers (SmartPractice) used in patch testing.46 In one case of aluminum-induced axillary dermatitis, a 28-year-old woman presented with eczema of the axillae, and subsequent patch testing revealed an allergy to aluminum chloride. The rash resolved upon cessation of use of an aluminum-containing deodorant.48

 

 

Aluminum has been reported to cause granulomatous dermatitis in the axillae. This reaction typically presents as red-brown, pruritic papules limited to the area in which deodorant was applied, with histopathology revealing epithelioid granulomas.49-51

Alum deodorants—considered a natural alternative—contain aluminum bound to potassium or ammonium in the form of a crystal or powder. Alum crystal deodorants have been reported to cause both a typical erythematous pruritic dermatitis as well as a granulomatous dermatitis with red-brown papules.52,53 The granulomatous dermatitis caused by either form of aluminum resolves with avoidance and use of topical steroids or topical tacrolimus.49,50,52,53

The diagnosis of aluminum ACD via patch testing may be identified with empty Finn Chambers, which are metallic aluminum, or with patch placement of aluminum chloride hexahydrate, though the former is only positive in patients with a strong allergy.54,55 In 2022, aluminum was named Allergen of the Year by the American Contact Dermatitis Society, with recommendations to conduct patch testing with aluminum chloride hexahydrate 10% rather than the traditional 2% to increase diagnostic yield.55 Additionally, it is recommended that aluminum be included in baseline patch testing for children due to the high prevalence of aluminum allergy in children and early exposure via childhood vaccines.54-56 In patients with aluminum allergy, providers may suggest purchasing aluminum-free deodorants or provide recipes for homemade deodorant that includes ingredients such as arrowroot powder, cornstarch, and diatomaceous earth.46

Nickel—Nickel is the most commonly identified contact allergen on patch testing yet an infrequent cause of axillary dermatitis. A case report from 2014 described axillary dermatitis in a woman that worsened during a positive patch test to nickel. Improvement was noted when the patient switched to titanium shaving razors.57 Nickel allergy also may present in the form of SCD. In one report, a woman developed dermatitis of the flexural areas, including the axillae, 3 months after undergoing a sterilization procedure in which nickel-containing tubal implants were placed.58 Patch testing revealed a positive reaction to nickel. The patient experienced complete resolution of the steroid-resistant dermatitis following removal of the implants via salpingectomy.58

Musicante_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Textile%20allergic%20contact%20dermatitis%20secondary%20to%20a%20deeply%20dyed%20blue%20sweater%20in%20a%20patient%20with%20positive%20patch%20test%20results%20to%20disperse%20blue%20106%2C%20disperse%20blue%20124%2C%20textile%20dye%20mix%2C%20formaldehyde%2C%20and%20methyldibromo%20glutaronitrile%2C%20among%20other%20allergens.%20The%20dermatitis%20involved%20the%20bilateral%20axillary%20rim%20and%20spared%20the%20vault.%3C%2Fp%3E

Textile Dye—In contrast to dermatitis caused by deodorants/antiperspirants, contact allergy to textile dyes presents as dermatitis involving the axillary borders but sparing the axillary vaults (Figures 2 and 3).10 Other potential presentations of textile dye dermatitis include erythema multiforme–like eruptions and erythematous wheal–type reactions.59 Textile dyes are classified as disperse vs nondisperse, with the majority of contact dermatoses caused by disperse dyes, specifically Disperse Orange 1, blue 106, and blue 124.60-62 Ryberg et al61 found that the axilla is one of the more common locations to be affected by textile dye allergy, particularly in women, which was further supported by Seidenari et al,63 who found that skin folds were affected in 27% of study participants allergic to textile dyes (N=437), a finding that is likely due to friction, sweat, and occlusion.62 In one case report of a patient with dermatitis caused by reactive dyes, the garment required 3 washes before the patient experienced resolution of dermatitis.64 For patients with textile dye dermatitis, mitigation strategies include washing clothing before wearing, especially for darkly dyed items; avoiding tight clothing; wearing garments made of cotton, wool, silk, or linen; and choosing light-colored garments.9,64,65

Musicante_3.jpg
%3Cp%3E%3Cstrong%3EFIGURE%203.%3C%2Fstrong%3E%20Allergic%20contact%20dermatitis%20of%20the%20axilla%20suspected%20to%20be%20secondary%20to%20black%20textile%20dyes.%20The%20dermatitis%20resolved%20completely%20with%20avoidance%20of%20tightly%20fitted%20black%20clothing.%3C%2Fp%3E

Axillary Dermatitis as a Manifestation of SCD and SDRIFE

Systemic contact dermatitis occurs when an individual who was previously sensitized to a particular allergen develops ACD of the skin with systemic exposure to that allergen or immunochemically related allergens. Exposure may occur via ingestion, inhalation, intravenous, intramuscular, and transepidermal routes.66 Systemic contact dermatitis manifests in a variety of ways, including focal flares at sites of prior contact dermatitis (recall reaction), vesicular hand dermatitis, intertriginous eruptions including axillary dermatitis, and generalized eruptions.67

Systemic contact dermatitis rarely involves systemic symptoms, and onset typically is within days of exposure. The 3 most common groups of allergens causing SCD are metals, medications, and plants and herbals.68 These allergens have all been reported to cause axillary dermatitis via SCD.58,69,70 Foods containing balsam of Peru that may lead to SCD include citrus, chocolate, tomato, and certain alcohols.70,71 Patients with a positive patch test to balsam of Peru may experience improvement of their dermatitis after reduction of balsam of Peru–rich foods from their diet.70 Metals implicated in SCD include mercury, nickel, and gold.72-74 Finally, PG ingestion also has been implicated in cases of SCD.37

 

 

Symmetrical drug-related intertriginous and flexural exanthema is another condition that presents as intertriginous dermatitis and differs from SCD in that the eruption does not require presensitization; there may be no known prior exposure to the agent causing dermatitis. Historically, SDRIFE was described as baboon syndrome because of its frequent involvement of the buttocks with diffuse, well-demarcated, erythematous dermatitis resembling that of a baboon. This term is no longer used due to its insensitive nature and incomplete depiction of SDRIFE, which can affect body sites other than the buttocks.68,75,76 Specific criteria to make this diagnosis include sharply demarcated and/or V-shaped erythema of the gluteal/perianal area, involvement of at least 1 other intertriginous or flexural region, symmetry of affected areas, and an absence of systemic symptoms.76 There also may be papules, pustules, and vesicles present in affected areas. Symmetrical drug-related intertriginous and flexural exanthema most often is caused by β-lactam antibiotics, but other associated drugs include chemotherapeutic agents, such as mitomycin C.76

Histopathology of both SCD and SDRIFE is variable and typically nonspecific, often revealing epidermal spongiosis and a perivascular mononuclear cell infiltrate with occasional neutrophils and eosinophils.76 A case of SCD to mercury presenting as intertriginous dermatitis demonstrated a leukocytoclastic vasculitis pattern on biopsy.77

Systemic contact dermatitis is diagnosed via a patch test, while SDRIFE typically has a negative patch test result and requires oral rechallenge testing, which reproduces the rash within hours.78,79

Additional Allergens Causing Axillary ACD

Although fragrance is the most common allergen in deodorants, other ingredients have been shown to cause axillary ACD (Table).80-90 In addition to these ingredients, allergens not previously mentioned that may be present in deodorants include lanolin, essential oils, and parabens.11 Methylisothiazolinone in laundry detergent also has been found to instigate ACD.91 Fragrances and preservatives in laundry detergents also may contribute to dermatitis.92

CT113001035_Table.jpg

Other products that have caused axillary contact dermatitis include topical exposure to medicaments including clindamycin,93 ethylenediamine in nystatin cream,94 methylprednisolone acetate95 and dipropylene glycol in a hydrocortisone lotion,96 wood dusts from tropical hardwoods,97 and tobacco.98

Management of ACD

The most effective strategy in the management of patients with contact dermatitis is avoidance of the offending agent. Additionally, clinicians may recommend the use of topical steroids and/or calcineurin inhibitors to hasten resolution.2

For patients with contact dermatitis, a clinician may recommend product substitutions with few potential allergens to use prior to patch testing. Patients with a fragrance allergy should look for products specifically labeled as “fragrance free” rather than “hypoallergenic” or “unscented,” as the latter two may still contain minimal amounts of fragrance.35 Patients should be educated on the functions of the allergens to which they are allergic so they may adequately avoid potential sources of contact.99 For suspected textile dye dermatitis, instructing patients to wash clothing before wearing and to avoid synthetic fabrics, dark dyes, and tightly fitted clothing may help.9,64,65

 

 

Differential Diagnosis

The differential diagnosis for axillary lesions is broad, including infectious, inflammatory, and autoimmune etiologies. Irritant contact dermatitis (ICD) presents similar to ACD, though it is more immediate in onsetand typically demonstrates symptoms of burning and stinging rather than pruritus. Although histopathology is not reliable in differentiating ICD and ACD, it has been shown that focal parakeratosis is associated with ACD, whereas necrotic epidermal keratinocytes are found in ICD.100

Intertrigo presents as large, erythematous, opposing patches or plaques confined to inguinal, submammary, axillary, and/or abdominal folds. Findings of beefy red erythema and peripheral satellite pustules may implicate presence of Candida, which can be identified with potassium hydroxide preparations.

Inverse psoriasis presents as sharply demarcated, erythematous, moist, smooth plaques or patches with minimal scale. The most common area of involvement is the inguinal folds, followed by the axillae, inframammary folds, perianal area, umbilicus, and retroauricular areas. Involvement of the elbows and knees or a positive family history of psoriasis may be useful knowledge in establishing the diagnosis. A biopsy may show dermal eosinophils, epidermal spongiosis, and focal serum in the scale, in addition to features of typical psoriasis plaques.101

Seborrheic dermatitis typically is an erythematous eruption, often with yellowish greasy scale. Simultaneous involvement of the face and scalp may be noted. Although typically a clinical diagnosis, biopsy demonstrates shoulder parakeratosis with follicular plugging and lymphocytic exocytosis.

Hailey-Hailey disease (also called benign familial pemphigus) is an autosomal-dominant genetic condition presenting as moist, malodorous, painful, vegetative plaques, patches, or scaly pustules in flexural areas, frequently with flaccid blisters. Lesions are provoked by traumatic stimuli. Onset occurs in the second to fourth decades and may improve with age. The diagnosis is confirmed by biopsy, which demonstrates acantholysis of the epidermis. The moist superficial patches of Hailey-Hailey disease help distinguish it from comparably drier Darier disease, the other acantholytic disease of the axillae.

Granular parakeratosis (also called hyperkeratotic flexural erythema) is an uncommon dermatosis most often observed in middle-aged women. It presents as red-brown keratotic papules coalescing into plaques, often with overlying scale in intertriginous areas. This disorder may be related to exposure to aluminum, a key component of antiperspirants.102 Diagnosis with a skin biopsy demonstrates granular parakeratosis.

Infections most commonly include erythrasma, tinea, and candidiasis. Erythrasma caused by Corynebacterium minutissimum may present in the axillae and/or groin with sharply demarcated, red-brown patches. Wood lamp examination reveals coral red fluorescence. Tinea corporis, a dermatophyte infection, may present as scaly erythematous plaques with advancing borders and central clearing. Fungal cultures and potassium hydroxide preparations are useful to confirm the diagnosis.

 

 

Pseudofolliculitis barbae most often is thought of as a condition affecting the beard in Black men, but it also may present in individuals of all races who shave the axillary and inguinal regions. Typical features include pruritic inflammatory papules and pustules with surrounding erythema and hyperpigmentation.

Fox-Fordyce disease is a disorder of the apocrine sweat glands that presents as several flesh-colored, perifollicular, monomorphic papules in the axillae. It typically is a disease of young females and also can involve the areola and vulva. Histopathology may show hyperkeratosis, irregular acanthosis, and dilated sweat glands.

Hidradenitis suppurativa is a chronic inflammatory condition that presents with multiple cysts; nodules; abscesses; sinus tract formation; and suppuration of the axillary, anogenital, and sometimes inframammary areas, typically at the onset of puberty. The diagnosis is best supported by history and physical examination, which may be notable for recurrent abscesses, draining tracts, double comedones, and ropelike scarring.

Extramammary Paget disease is a rare malignancy affecting apocrine gland–bearing areas, including axillary and genital regions. It most commonly presents as a unilateral or asymmetric, scaly, erythematous plaque. Histopathology demonstrates Paget cells with abundant clear cytoplasm and pleomorphic nuclei, typically grouped in the lower portion of the epidermis.

Final Thoughts

Axillary dermatoses often can be challenging to diagnose given the range of pathologies that can present in intertriginous areas. Allergic contact dermatitis is a common culprit due to unique anatomical considerations and self-care practices, including shaving/hair removal; use of deodorants, antiperspirants, bodywashes, and clothing; and frictional and moisture influences. The most likely offender among contact allergens is fragrance, but other possibilities to consider include PG, preservatives, aluminum, nickel, and textile dyes. Albeit less common, systemic exposure to allergens may result in SCD and SDRIFE with a rash in intertriginous zones, including the axillae. Additionally, other infectious, inflammatory, and autoimmune etiologies should be considered and ruled out.

Patch testing is the most reliable method to diagnose suspected ACD. Once confirmed, management includes the use of topical steroids and avoidance of the causative agent. Additionally, patients should be informed of the American Contact Dermatitis Society Contact Allergen Management Program (https://www.contactderm.org/patient-support/camp-access), which provides patients with useful information on products that are safe to use based on their patch testing results.

References
  1. Alinaghi F, Bennike NH, Egeberg A, et al. Prevalence of contact allergy in the general population: a systematic review and meta-analysis. Contact Dermatitis. 2019;80:77-85.
  2. Brar KK. A review of contact dermatitis. Ann Allergy Asthma Immunol. 2021;126:32-39.
  3. Evans RL, Marriott RE, Harker M. Axillary skin: biology and care. Int J Cosmet Sci. 2012;34:389-395.
  4. Watkinson A, Lee RS, Moore AE, et al. Is the axilla a distinct skin phenotype? Int J Cosmet Sci. 2007;29:60.
  5. Wu JQ, Kilpatrick-Liverman L. Characterizing the composition of underarm and forearm skin using confocal raman spectroscopy. Int J Cosmet Sci. 2011;33:257-262.
  6. Marti VP, Lee RS, Moore AE, et al. Effect of shaving on axillary stratum corneum. Int J Cosmet Sci. 2003;25:193-198.
  7. Turner GA, Moore AE, Marti VPJ, et al. Impact of shaving and anti-perspirant use on the axillary vault. Int J Cosmet Sci. 2007;29:31-38.
  8. Zhai H, Maibach HI. Skin occlusion and irritant and allergic contact dermatitis: an overview. Contact Dermatitis. 2001;44:201-206.
  9. Lazarov A. Textile dermatitis in patients with contact sensitization in Israel: a 4-year prospective study. J Eur Acad Dermatol Venereol. 2004;18:531-537.
  10. Nelson JL, Mowad CM. Allergic contact dermatitis: patch testing beyond the TRUE Test. J Clin Aesthet Dermatol. 2010;3:36-41.
  11. Zirwas MJ, Moennich J. Antiperspirant and deodorant allergy: diagnosis and management. J Clin Aesthet Dermatol. 2008;1:38-43.
  12. DeKoven JG, Warshaw EM, Reeder MJ, et al. North American Contact Dermatitis Group Patch Test Results: 2019-2020. Dermatitis. 2023;34:90-104.
  13. Eiermann HJ, Larsen W, Maibach HI, et al. Prospective study of cosmetic reactions: 1977-1980. North American Contact Dermatitis Group. J Am Acad Dermatol. 1982;6:909-917.
  14. González-Muñoz P, Conde-Salazar L, Vañó-Galván S. Allergic contact dermatitis caused by cosmetic products. Actas Dermosifiliogr. 2014;105:822-832.
  15. Gerberick GF, Robinson MK, Felter SP, et al. Understanding fragrance allergy using an exposure-based risk assessment approach. Contact Dermatitis. 2001;45:333-340.
  16. Heisterberg MV, Menne T, Andersen KE, et al. Deodorants are the leading cause of allergic contact dermatitis to fragrance ingredients. Contact Dermatitis. 2011;64:258-264.
  17. Johansen JD, Andersen TF, Kjoller M, et al. Identification of risk products for fragrance contact allergy: a case-referent study based on patients’ histories. Am J Contact Dermat. 1998;9:80-86.
  18. Edman B. The influence of shaving method on perfume allergy. Contact Dermatitis. 1994;31:291-292.
  19. Hamza M, Tohid H, Maibach H. Shaving effects on percutaneous penetration: clinical implications. Cutan Ocul Toxicol. 2015;34:335-343.
  20. Geier J, Uter W, Lessmann H, et al. Fragrance mix I and II: results of breakdown tests. Flavour Fragr J. 2015;30:264-274.
  21. Handley J, Burrows D. Allergic contact dermatitis from the synthetic fragrances Lyral and acetyl cedrene in separate underarm deodorant preparations. Contact Dermatitis. 1994;31:288-290.
  22. Hendriks SA, Bousema MT, van Ginkel CJ. Allergic contact dermatitis from the fragrance ingredient Lyral in underarm deodorant. Contact Dermatitis. 1999;41:119.
  23. Jacob SE. Allergic contact dermatitis from lyral in an aerosol deodorant. Dermatitis. 2008;19:216-217.
  24. Gilpin S, Maibach H. Allergic contact dermatitis caused by farnesol: clinical relevance. Cutan Ocul Toxicol. 2010;29:278-287.
  25. Goossens A, Merckx L. Allergic contact dermatitis from farnesol in a deodorant. Contact Dermatitis. 1997;37:179-180.
  26. Schnuch A, Uter W, Geier J, et al. Contact allergy to farnesol in 2021 consecutively patch tested patients. Results of the IVDK. Contact Dermatitis. 2004;50:117-121.
  27. Uter W, Geier J, Schnuch A, et al. Patch test results with patients’ own perfumes, deodorants and shaving lotions: results of the IVDK 1998–2002. J Eur Acad Dermatol Venereol. 2007;21:374-379.
  28. Dittmar D, Schuttelaar MLA. Contact sensitization to hydroperoxides of limonene and linalool: results of consecutive patch testing and clinical relevance. Contact Dermatitis. 2019;80:101-109.
  29. Yazar K, Johnsson S, Lind M-L, et al. Preservatives and fragrances in selected consumer-available cosmetics and detergents. Contact Dermatitis. 2011;64:265-272.
  30. Isaksson M, Karlberg A-T, Nilsson U. Allergic contact dermatitis caused by oxidized linalool in a deodorant. Contact Dermatitis. 2019;81:213-214.
  31. Chen J, Yi Z, Sun R, et al. Analysis of fragrance allergens in personal care products, toys, and water samples: a review. J AOAC Int. 2022;105:396-412.
  32. Larsen WG. Perfume dermatitis. J Am Acad Dermatol. 1985;12:1-9.
  33. Pincelli C, Magni R, Motolese A. Pigmented contact dermatitis from deodorant. Contact Dermatitis. 1993;28:305-306.
  34. Kwong HL, Lim SPR. Pigmented contact dermatitis in the axillae caused by hydroperoxides of limonene. JAAD Case Reports. 2020;6:476-478.
  35. Marks J, Anderson B, DeLeo V. Contact and Occupational Dermatology. 4th ed. Jaypee; 2016.
  36. Johansen JD. Fragrance contact allergy: a clinical review. Am J Clin Dermatol. 2003;4:789-798.
  37. McGowan MA, Scheman A, Jacob SE. Propylene glycol in contact dermatitis: a systematic review. Dermatitis. 2018;29:6-12.
  38. Fiume MM, Bergfeld WF, Belsito DV, et al. Safety assessment of propylene glycol, tripropylene glycol, and PPGs as used in cosmetics. Int J Toxicol. 2012;31(5 suppl):245S-260S.
  39. Farrar CW, Bell HK, King CM. Allergic contact dermatitis from propylene glycol in Efudix cream. Contact Dermatitis. 2003;48:345.
  40. Friedman ES, Friedman PM, Cohen DE, et al. Allergic contact dermatitis to topical minoxidil solution: etiology and treatment. J Am Acad Dermatol. 2002;46:309-312.
  41. Lessmann H, Schnuch A, Geier J, et al. Skin-sensitizing and irritant properties of propylene glycol. Contact Dermatitis. 2005;53:247-259.
  42. Agren-Jonsson S, Magnusson B. Sensitization to propantheline bromide, trichlorocarbanilide and propylene glycol in an antiperspirant. Contact Dermatitis. 1976;2:79-80.
  43. Catanzaro JM, Smith JG Jr. Propylene glycol dermatitis. J Am Acad Dermatol. 1991;24:90-95.
  44. Jacob SE, Scheman A, McGowan MA. Propylene glycol. Dermatitis. 2018;29:3-5.
  45. Carlson S, Gipson K, Nedorost S. Relevance of doubtful (“equivocal”) late patch-test readings. Dermatitis. 2010;21:102-108.
  46. Kullberg SA, Ward JM, Liou YL, et al. Cutaneous reactions to aluminum. Dermatitis. 2020;31:335-349.
  47. Benohanian A. Antiperspirants and deodorants. Clin Dermatol. 2001;19:398-405.
  48. Garg S, Loghdey S, Gawkrodger DJ. Allergic contact dermatitis from aluminum in deodorants. Contact Dermatitis. 2010;62:57-58.
  49. Montemarano AD, Sau P, Johnson FB, et al. Cutaneous granulomas caused by an aluminum-zirconium complex: an ingredient of antiperspirants. J Am Acad Dermatol. 1997;37:496-498.
  50. Rubin L, Slepyan AH, Weber LF, et al. Granulomas of the axillae caused by deodorants. JAMA. 1956;162:953-955.
  51. Williams S, Freemont AJ. Aerosol antiperspirants and axillary granulomata. Br Med J (Clin Res Ed). 1984;288:1651-1652.
  52. Gallego H, Lewis EJ, Crutchfield CE 3rd. Crystal deodorant dermatitis: irritant dermatitis to alum-containing deodorant. Cutis. 1999;64:65-66.
  53. Leventhal JS, Farhadian JA, Miller KE, et al. Crystal deodorant-induced axillary granulomatous dermatitis. Int J Dermatol. 2014;53:e59-e60.
  54. Siemund I, Dahlin J, Hindsén M, et al. Contact allergy to two aluminum salts in consecutively patch-tested dermatitis patients. Dermatitis. 2022;3:31-35.
  55. Bruze M, Netterlid E, Siemund I. Aluminum-allergen of the year 2022. Dermatitis. 2022;33:10-15.
  56. Goiset A, Darrigade A-S, Labrèze C, et al. Aluminum sensitization in a French paediatric patch test population. Contact Dermatitis. 2018;79:382-383.
  57. Admani S, Matiz C, Jacob SE. Nickel allergy—a potential cause of razor dermatitis. Pediatr Dermatol. 2014;31:392-393.
  58. Bibas N, Lassere J, Paul C, et al. Nickel-induced systemic contact dermatitis and intratubal implants: the baboon syndrome revisited. Dermatitis. 2013;24:35-36.
  59. Seidenari S, Manzini BM, Ddanese P. Contact sensitization to textile dyes: description of 100 subjects. Contact Dermatitis. 1991;24:253-258.
  60. Hatch KL, Maibach HI. Textile dye allergic contact dermatitis prevalence. Contact Dermatitis. 2000;42:187-195.
  61. Ryberg K, Isaksson M, Gruvberger B, et al. Contact allergy to textile dyes in southern Sweden. Contact Dermatitis. 2006;54:313-321.
  62. Pratt M, Taraska V. Disperse blue dyes 106 and 124 are common causes of textile dermatitis and should serve as screening allergens for this condition. Dermatitis. 2000;11:30-41.
  63. Seidenari S, Giusti F, Massone F, et al. Sensitization to disperse dyes in a patch test population over a five-year period. Am J Contact Dermat. 2002;13:101-107.
  64. Moreau L, Goossens A. Allergic contact dermatitis associated with reactive dyes in a dark garment: a case report. Contact Dermatitis. 2005;53:150-154.
  65. Svedman C, Engfeldt M, Malinauskiene L. Textile contact dermatitis: how fabrics can induce dermatitis. Curr Treat Options Allergy. 2019;6:103-111.
  66. Jacob SE, Zapolanski T. Systemic contact dermatitis. Dermatitis. 2008;19:9-15.
  67. Hindsén M, Bruze M, Christensen OB. Flare-up reactions after oral challenge with nickel in relation to challenge dose and intensity and time of previous patch test reactions. J Am Acad Dermatol. 2001;44:616-623.
  68. Winnicki M, Shear NH. A systematic approach to systemic contact dermatitis and symmetric drug-related intertriginous and flexural exanthema (SDRIFE): a closer look at these conditions and an approach to intertriginous eruptions. Am J Clin Dermatol. 2011;12:171-180.
  69. Kalita BJ, Das S, Dutta B. Itraconazole-induced symmetrical drug-related intertriginous and flexural exanthema (SDRIFE): a rare occurrence. Int J Dermatol. 2020;59:e419-e421.
  70. Salam TN, Fowler JF Jr. Balsam-related systemic contact dermatitis. J Am Acad Dermatol. 2001;45:377-381.
  71. Ramachandran V, Cline A, Summey B, et al. Systemic contact dermatitis related to alcoholic beverage consumption. Dermatol Online J. 2019;25:13030/qt3zg853qv.
  72. Moreno-Ramírez D, García-Bravo B, Pichardo AR, et al. Baboon syndrome in childhood: easy to avoid, easy to diagnose, but the problem continues. Pediatr Dermatol. 2004;21:250-253.
  73. Dou X, Liu L-L, Zhu X-J. Nickel-elicited systemic contact dermatitis. Contact Dermatitis. 2003;48:126-129.
  74. Möller H, Ohlsson K, Linder C, et al. The flare-up reactions after systemic provocation in contact allergy to nickel and gold. Contact Dermatitis. 1999;40:200-204.
  75. Andersen KE, Hjorth N, Menné T. The baboon syndrome: systemically-induced allergic contact dermatitis. Contact Dermatitis. 1984;10:97-100.
  76. Häusermann P, Harr T, Bircher AJ. Baboon syndrome resulting from systemic drugs: is there strife between SDRIFE and allergic contact dermatitis syndrome? Contact Dermatitis. 2004;51:297-310.
  77. Tan MG, Pratt MD, Burns BF, et al. Baboon syndrome from mercury showing leukocytoclastic vasculitis on biopsy. Contact Dermatitis. 2020;83:415-417.
  78. Handisurya A, Stingl G, Wöhrl S. SDRIFE (baboon syndrome) induced by penicillin. Clin Exp Dermatol. 2009;34:355-357.
  79. Akay BN, Sanli H. Symmetrical drug-related intertriginous and flexural exanthem due to oral risperidone. Pediatr Dermatol. 2009;26:214-216.
  80. Amaro C, Santos R, Cardoso J. Contact allergy to methylisothiazolinone in a deodorant. Contact Dermatitis. 2011;64:298-299.
  81. Goh CL. Dermatitis from chlorphenesin in a deodorant. Contact Dermatitis. 1987;16:287.
  82. Taghipour K, Tatnall F, Orton D. Allergic axillary dermatitis due to hydrogenated castor oil in a deodorant. Contact Dermatitis. 2008;58:168-169.
  83. Sheu M, Simpson EL, Law S V, et al. Allergic contact dermatitis from a natural deodorant: a report of 4 cases associated with lichen acid mix allergy. J Am Acad Dermatol. 2006;55:332-337.
  84. Pastor-Nieto M-A, Gatica-Ortega M-E, Alcántara-Nicolás F-D-A, et al. Allergic contact dermatitis resulting from cetyl PEG/PPG-10/1 dimethicone in a deodorant cream. Contact Dermatitis. 2018;78:236-239.
  85. Corazza M, Lombardi AR, Virgili A. Non-eczematous urticarioid allergic contact dermatitis due to Eumulgin L in a deodorant. Contact Dermatitis. 1997;36:159-160.
  86. van Ketel WG. Allergic contact dermatitis from propellants in deodorant sprays in combination with allergy to ethyl chloride. Contact Dermatitis. 1976;2:115-119.
  87. Shmunes E, Levy EJ. Quaternary ammonium compound contact dermatitis from a deodorant. Arch Dermatol. 1972;105:91-93.
  88. Bruze M, Johansen JD, Andersen KE, et al. Deodorants: an experimental provocation study with cinnamic aldehyde. J Am Acad Dermatol. 2003;48:194-200.
  89. Hann S, Hughes TM, Stone NM. Flexural allergic contact dermatitis to benzalkonium chloride in antiseptic bath oil. Br J Dermatol. 2007;157:795-798.
  90. Aeling JL, Panagotacos PJ, Andreozzi RJ. Allergic contact dermatitis to vitamin E aerosol deodorant. Arch Dermatol. 1973;108:579-580.
  91. Cotton CH, Duah CG, Matiz C. Allergic contact dermatitis due to methylisothiazolinone in a young girl’s laundry detergent. Pediatr Dermatol. 2017;34:486-487.
  92. Magnano M, Silvani S, Vincenzi C, et al. Contact allergens and irritants in household washing and cleaning products. Contact Dermatitis. 2009;61:337-341.
  93. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. Contact Dermatitis. 2020;82:313-314.
  94. Iammatteo M, Akenroye A, Jariwala S, et al. Severe contact dermatitis due to ethylenediamine dihydrochloride in nystatin cream. J Allergy Clin Immunol Pract. 2017;5:1448-1450.
  95. Coskey RJ, Bryan HG. Contact dermatitis due to methylprednisolone. JAMA. 1967;199:136.
  96. Peterson MY, Han J, Warshaw EM. Allergic contact dermatitis from dipropylene glycol in hydrocortisone lotion. Contact Dermatitis. 2022;87:112-114.
  97. Ferreira O, Cruz MJ, Mota A, et al. Erythema multiforme-like lesions revealing allergic contact dermatitis to exotic woods. Cutan Ocul Toxicol. 2012;31:61-63.
  98. Abraham NF, Feldman SR, Vallejos Q, et al. Contact dermatitis in tobacco farmworkers. Contact Dermatitis. 2007;57:40-43.
  99. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. J Am Acad Dermatol. 2016;74:1043-1054.
  100. Frings VG, Böer-Auer A, Breuer K. Histomorphology and immunophenotype of eczematous skin lesions revisited-skinbiopsies are not reliable in differentiating allergic contact dermatitis, irritant contact dermatitis, and atopic dermatitis. Am J Dermatopathol. 2018;40:7-16.
  101. Knabel M, Mudaliar K. Histopathologic features of inverse psoriasis. J Cutan Pathol. 2022;49:246-251.
  102. Fujii M, Kishibe M, Honma M, et al. Aluminum chloride-induced apoptosis leads to keratinization arrest and granular parakeratosis. Am J Dermatopathol. 2020;42:756-761.
References
  1. Alinaghi F, Bennike NH, Egeberg A, et al. Prevalence of contact allergy in the general population: a systematic review and meta-analysis. Contact Dermatitis. 2019;80:77-85.
  2. Brar KK. A review of contact dermatitis. Ann Allergy Asthma Immunol. 2021;126:32-39.
  3. Evans RL, Marriott RE, Harker M. Axillary skin: biology and care. Int J Cosmet Sci. 2012;34:389-395.
  4. Watkinson A, Lee RS, Moore AE, et al. Is the axilla a distinct skin phenotype? Int J Cosmet Sci. 2007;29:60.
  5. Wu JQ, Kilpatrick-Liverman L. Characterizing the composition of underarm and forearm skin using confocal raman spectroscopy. Int J Cosmet Sci. 2011;33:257-262.
  6. Marti VP, Lee RS, Moore AE, et al. Effect of shaving on axillary stratum corneum. Int J Cosmet Sci. 2003;25:193-198.
  7. Turner GA, Moore AE, Marti VPJ, et al. Impact of shaving and anti-perspirant use on the axillary vault. Int J Cosmet Sci. 2007;29:31-38.
  8. Zhai H, Maibach HI. Skin occlusion and irritant and allergic contact dermatitis: an overview. Contact Dermatitis. 2001;44:201-206.
  9. Lazarov A. Textile dermatitis in patients with contact sensitization in Israel: a 4-year prospective study. J Eur Acad Dermatol Venereol. 2004;18:531-537.
  10. Nelson JL, Mowad CM. Allergic contact dermatitis: patch testing beyond the TRUE Test. J Clin Aesthet Dermatol. 2010;3:36-41.
  11. Zirwas MJ, Moennich J. Antiperspirant and deodorant allergy: diagnosis and management. J Clin Aesthet Dermatol. 2008;1:38-43.
  12. DeKoven JG, Warshaw EM, Reeder MJ, et al. North American Contact Dermatitis Group Patch Test Results: 2019-2020. Dermatitis. 2023;34:90-104.
  13. Eiermann HJ, Larsen W, Maibach HI, et al. Prospective study of cosmetic reactions: 1977-1980. North American Contact Dermatitis Group. J Am Acad Dermatol. 1982;6:909-917.
  14. González-Muñoz P, Conde-Salazar L, Vañó-Galván S. Allergic contact dermatitis caused by cosmetic products. Actas Dermosifiliogr. 2014;105:822-832.
  15. Gerberick GF, Robinson MK, Felter SP, et al. Understanding fragrance allergy using an exposure-based risk assessment approach. Contact Dermatitis. 2001;45:333-340.
  16. Heisterberg MV, Menne T, Andersen KE, et al. Deodorants are the leading cause of allergic contact dermatitis to fragrance ingredients. Contact Dermatitis. 2011;64:258-264.
  17. Johansen JD, Andersen TF, Kjoller M, et al. Identification of risk products for fragrance contact allergy: a case-referent study based on patients’ histories. Am J Contact Dermat. 1998;9:80-86.
  18. Edman B. The influence of shaving method on perfume allergy. Contact Dermatitis. 1994;31:291-292.
  19. Hamza M, Tohid H, Maibach H. Shaving effects on percutaneous penetration: clinical implications. Cutan Ocul Toxicol. 2015;34:335-343.
  20. Geier J, Uter W, Lessmann H, et al. Fragrance mix I and II: results of breakdown tests. Flavour Fragr J. 2015;30:264-274.
  21. Handley J, Burrows D. Allergic contact dermatitis from the synthetic fragrances Lyral and acetyl cedrene in separate underarm deodorant preparations. Contact Dermatitis. 1994;31:288-290.
  22. Hendriks SA, Bousema MT, van Ginkel CJ. Allergic contact dermatitis from the fragrance ingredient Lyral in underarm deodorant. Contact Dermatitis. 1999;41:119.
  23. Jacob SE. Allergic contact dermatitis from lyral in an aerosol deodorant. Dermatitis. 2008;19:216-217.
  24. Gilpin S, Maibach H. Allergic contact dermatitis caused by farnesol: clinical relevance. Cutan Ocul Toxicol. 2010;29:278-287.
  25. Goossens A, Merckx L. Allergic contact dermatitis from farnesol in a deodorant. Contact Dermatitis. 1997;37:179-180.
  26. Schnuch A, Uter W, Geier J, et al. Contact allergy to farnesol in 2021 consecutively patch tested patients. Results of the IVDK. Contact Dermatitis. 2004;50:117-121.
  27. Uter W, Geier J, Schnuch A, et al. Patch test results with patients’ own perfumes, deodorants and shaving lotions: results of the IVDK 1998–2002. J Eur Acad Dermatol Venereol. 2007;21:374-379.
  28. Dittmar D, Schuttelaar MLA. Contact sensitization to hydroperoxides of limonene and linalool: results of consecutive patch testing and clinical relevance. Contact Dermatitis. 2019;80:101-109.
  29. Yazar K, Johnsson S, Lind M-L, et al. Preservatives and fragrances in selected consumer-available cosmetics and detergents. Contact Dermatitis. 2011;64:265-272.
  30. Isaksson M, Karlberg A-T, Nilsson U. Allergic contact dermatitis caused by oxidized linalool in a deodorant. Contact Dermatitis. 2019;81:213-214.
  31. Chen J, Yi Z, Sun R, et al. Analysis of fragrance allergens in personal care products, toys, and water samples: a review. J AOAC Int. 2022;105:396-412.
  32. Larsen WG. Perfume dermatitis. J Am Acad Dermatol. 1985;12:1-9.
  33. Pincelli C, Magni R, Motolese A. Pigmented contact dermatitis from deodorant. Contact Dermatitis. 1993;28:305-306.
  34. Kwong HL, Lim SPR. Pigmented contact dermatitis in the axillae caused by hydroperoxides of limonene. JAAD Case Reports. 2020;6:476-478.
  35. Marks J, Anderson B, DeLeo V. Contact and Occupational Dermatology. 4th ed. Jaypee; 2016.
  36. Johansen JD. Fragrance contact allergy: a clinical review. Am J Clin Dermatol. 2003;4:789-798.
  37. McGowan MA, Scheman A, Jacob SE. Propylene glycol in contact dermatitis: a systematic review. Dermatitis. 2018;29:6-12.
  38. Fiume MM, Bergfeld WF, Belsito DV, et al. Safety assessment of propylene glycol, tripropylene glycol, and PPGs as used in cosmetics. Int J Toxicol. 2012;31(5 suppl):245S-260S.
  39. Farrar CW, Bell HK, King CM. Allergic contact dermatitis from propylene glycol in Efudix cream. Contact Dermatitis. 2003;48:345.
  40. Friedman ES, Friedman PM, Cohen DE, et al. Allergic contact dermatitis to topical minoxidil solution: etiology and treatment. J Am Acad Dermatol. 2002;46:309-312.
  41. Lessmann H, Schnuch A, Geier J, et al. Skin-sensitizing and irritant properties of propylene glycol. Contact Dermatitis. 2005;53:247-259.
  42. Agren-Jonsson S, Magnusson B. Sensitization to propantheline bromide, trichlorocarbanilide and propylene glycol in an antiperspirant. Contact Dermatitis. 1976;2:79-80.
  43. Catanzaro JM, Smith JG Jr. Propylene glycol dermatitis. J Am Acad Dermatol. 1991;24:90-95.
  44. Jacob SE, Scheman A, McGowan MA. Propylene glycol. Dermatitis. 2018;29:3-5.
  45. Carlson S, Gipson K, Nedorost S. Relevance of doubtful (“equivocal”) late patch-test readings. Dermatitis. 2010;21:102-108.
  46. Kullberg SA, Ward JM, Liou YL, et al. Cutaneous reactions to aluminum. Dermatitis. 2020;31:335-349.
  47. Benohanian A. Antiperspirants and deodorants. Clin Dermatol. 2001;19:398-405.
  48. Garg S, Loghdey S, Gawkrodger DJ. Allergic contact dermatitis from aluminum in deodorants. Contact Dermatitis. 2010;62:57-58.
  49. Montemarano AD, Sau P, Johnson FB, et al. Cutaneous granulomas caused by an aluminum-zirconium complex: an ingredient of antiperspirants. J Am Acad Dermatol. 1997;37:496-498.
  50. Rubin L, Slepyan AH, Weber LF, et al. Granulomas of the axillae caused by deodorants. JAMA. 1956;162:953-955.
  51. Williams S, Freemont AJ. Aerosol antiperspirants and axillary granulomata. Br Med J (Clin Res Ed). 1984;288:1651-1652.
  52. Gallego H, Lewis EJ, Crutchfield CE 3rd. Crystal deodorant dermatitis: irritant dermatitis to alum-containing deodorant. Cutis. 1999;64:65-66.
  53. Leventhal JS, Farhadian JA, Miller KE, et al. Crystal deodorant-induced axillary granulomatous dermatitis. Int J Dermatol. 2014;53:e59-e60.
  54. Siemund I, Dahlin J, Hindsén M, et al. Contact allergy to two aluminum salts in consecutively patch-tested dermatitis patients. Dermatitis. 2022;3:31-35.
  55. Bruze M, Netterlid E, Siemund I. Aluminum-allergen of the year 2022. Dermatitis. 2022;33:10-15.
  56. Goiset A, Darrigade A-S, Labrèze C, et al. Aluminum sensitization in a French paediatric patch test population. Contact Dermatitis. 2018;79:382-383.
  57. Admani S, Matiz C, Jacob SE. Nickel allergy—a potential cause of razor dermatitis. Pediatr Dermatol. 2014;31:392-393.
  58. Bibas N, Lassere J, Paul C, et al. Nickel-induced systemic contact dermatitis and intratubal implants: the baboon syndrome revisited. Dermatitis. 2013;24:35-36.
  59. Seidenari S, Manzini BM, Ddanese P. Contact sensitization to textile dyes: description of 100 subjects. Contact Dermatitis. 1991;24:253-258.
  60. Hatch KL, Maibach HI. Textile dye allergic contact dermatitis prevalence. Contact Dermatitis. 2000;42:187-195.
  61. Ryberg K, Isaksson M, Gruvberger B, et al. Contact allergy to textile dyes in southern Sweden. Contact Dermatitis. 2006;54:313-321.
  62. Pratt M, Taraska V. Disperse blue dyes 106 and 124 are common causes of textile dermatitis and should serve as screening allergens for this condition. Dermatitis. 2000;11:30-41.
  63. Seidenari S, Giusti F, Massone F, et al. Sensitization to disperse dyes in a patch test population over a five-year period. Am J Contact Dermat. 2002;13:101-107.
  64. Moreau L, Goossens A. Allergic contact dermatitis associated with reactive dyes in a dark garment: a case report. Contact Dermatitis. 2005;53:150-154.
  65. Svedman C, Engfeldt M, Malinauskiene L. Textile contact dermatitis: how fabrics can induce dermatitis. Curr Treat Options Allergy. 2019;6:103-111.
  66. Jacob SE, Zapolanski T. Systemic contact dermatitis. Dermatitis. 2008;19:9-15.
  67. Hindsén M, Bruze M, Christensen OB. Flare-up reactions after oral challenge with nickel in relation to challenge dose and intensity and time of previous patch test reactions. J Am Acad Dermatol. 2001;44:616-623.
  68. Winnicki M, Shear NH. A systematic approach to systemic contact dermatitis and symmetric drug-related intertriginous and flexural exanthema (SDRIFE): a closer look at these conditions and an approach to intertriginous eruptions. Am J Clin Dermatol. 2011;12:171-180.
  69. Kalita BJ, Das S, Dutta B. Itraconazole-induced symmetrical drug-related intertriginous and flexural exanthema (SDRIFE): a rare occurrence. Int J Dermatol. 2020;59:e419-e421.
  70. Salam TN, Fowler JF Jr. Balsam-related systemic contact dermatitis. J Am Acad Dermatol. 2001;45:377-381.
  71. Ramachandran V, Cline A, Summey B, et al. Systemic contact dermatitis related to alcoholic beverage consumption. Dermatol Online J. 2019;25:13030/qt3zg853qv.
  72. Moreno-Ramírez D, García-Bravo B, Pichardo AR, et al. Baboon syndrome in childhood: easy to avoid, easy to diagnose, but the problem continues. Pediatr Dermatol. 2004;21:250-253.
  73. Dou X, Liu L-L, Zhu X-J. Nickel-elicited systemic contact dermatitis. Contact Dermatitis. 2003;48:126-129.
  74. Möller H, Ohlsson K, Linder C, et al. The flare-up reactions after systemic provocation in contact allergy to nickel and gold. Contact Dermatitis. 1999;40:200-204.
  75. Andersen KE, Hjorth N, Menné T. The baboon syndrome: systemically-induced allergic contact dermatitis. Contact Dermatitis. 1984;10:97-100.
  76. Häusermann P, Harr T, Bircher AJ. Baboon syndrome resulting from systemic drugs: is there strife between SDRIFE and allergic contact dermatitis syndrome? Contact Dermatitis. 2004;51:297-310.
  77. Tan MG, Pratt MD, Burns BF, et al. Baboon syndrome from mercury showing leukocytoclastic vasculitis on biopsy. Contact Dermatitis. 2020;83:415-417.
  78. Handisurya A, Stingl G, Wöhrl S. SDRIFE (baboon syndrome) induced by penicillin. Clin Exp Dermatol. 2009;34:355-357.
  79. Akay BN, Sanli H. Symmetrical drug-related intertriginous and flexural exanthem due to oral risperidone. Pediatr Dermatol. 2009;26:214-216.
  80. Amaro C, Santos R, Cardoso J. Contact allergy to methylisothiazolinone in a deodorant. Contact Dermatitis. 2011;64:298-299.
  81. Goh CL. Dermatitis from chlorphenesin in a deodorant. Contact Dermatitis. 1987;16:287.
  82. Taghipour K, Tatnall F, Orton D. Allergic axillary dermatitis due to hydrogenated castor oil in a deodorant. Contact Dermatitis. 2008;58:168-169.
  83. Sheu M, Simpson EL, Law S V, et al. Allergic contact dermatitis from a natural deodorant: a report of 4 cases associated with lichen acid mix allergy. J Am Acad Dermatol. 2006;55:332-337.
  84. Pastor-Nieto M-A, Gatica-Ortega M-E, Alcántara-Nicolás F-D-A, et al. Allergic contact dermatitis resulting from cetyl PEG/PPG-10/1 dimethicone in a deodorant cream. Contact Dermatitis. 2018;78:236-239.
  85. Corazza M, Lombardi AR, Virgili A. Non-eczematous urticarioid allergic contact dermatitis due to Eumulgin L in a deodorant. Contact Dermatitis. 1997;36:159-160.
  86. van Ketel WG. Allergic contact dermatitis from propellants in deodorant sprays in combination with allergy to ethyl chloride. Contact Dermatitis. 1976;2:115-119.
  87. Shmunes E, Levy EJ. Quaternary ammonium compound contact dermatitis from a deodorant. Arch Dermatol. 1972;105:91-93.
  88. Bruze M, Johansen JD, Andersen KE, et al. Deodorants: an experimental provocation study with cinnamic aldehyde. J Am Acad Dermatol. 2003;48:194-200.
  89. Hann S, Hughes TM, Stone NM. Flexural allergic contact dermatitis to benzalkonium chloride in antiseptic bath oil. Br J Dermatol. 2007;157:795-798.
  90. Aeling JL, Panagotacos PJ, Andreozzi RJ. Allergic contact dermatitis to vitamin E aerosol deodorant. Arch Dermatol. 1973;108:579-580.
  91. Cotton CH, Duah CG, Matiz C. Allergic contact dermatitis due to methylisothiazolinone in a young girl’s laundry detergent. Pediatr Dermatol. 2017;34:486-487.
  92. Magnano M, Silvani S, Vincenzi C, et al. Contact allergens and irritants in household washing and cleaning products. Contact Dermatitis. 2009;61:337-341.
  93. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. Contact Dermatitis. 2020;82:313-314.
  94. Iammatteo M, Akenroye A, Jariwala S, et al. Severe contact dermatitis due to ethylenediamine dihydrochloride in nystatin cream. J Allergy Clin Immunol Pract. 2017;5:1448-1450.
  95. Coskey RJ, Bryan HG. Contact dermatitis due to methylprednisolone. JAMA. 1967;199:136.
  96. Peterson MY, Han J, Warshaw EM. Allergic contact dermatitis from dipropylene glycol in hydrocortisone lotion. Contact Dermatitis. 2022;87:112-114.
  97. Ferreira O, Cruz MJ, Mota A, et al. Erythema multiforme-like lesions revealing allergic contact dermatitis to exotic woods. Cutan Ocul Toxicol. 2012;31:61-63.
  98. Abraham NF, Feldman SR, Vallejos Q, et al. Contact dermatitis in tobacco farmworkers. Contact Dermatitis. 2007;57:40-43.
  99. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. J Am Acad Dermatol. 2016;74:1043-1054.
  100. Frings VG, Böer-Auer A, Breuer K. Histomorphology and immunophenotype of eczematous skin lesions revisited-skinbiopsies are not reliable in differentiating allergic contact dermatitis, irritant contact dermatitis, and atopic dermatitis. Am J Dermatopathol. 2018;40:7-16.
  101. Knabel M, Mudaliar K. Histopathologic features of inverse psoriasis. J Cutan Pathol. 2022;49:246-251.
  102. Fujii M, Kishibe M, Honma M, et al. Aluminum chloride-induced apoptosis leads to keratinization arrest and granular parakeratosis. Am J Dermatopathol. 2020;42:756-761.
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
35-42
Page Number
35-42
Publications
Publications
Topics
Article Type
Display Headline
Axillary Contact Dermatitis: An Update on Potential Allergens and Management
Display Headline
Axillary Contact Dermatitis: An Update on Potential Allergens and Management
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Musicante</fileName> <TBEID>0C02EFC3.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EFC3</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Musicante</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240108T080648</firstPublished> <LastPublished>20240108T080648</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240108T080648</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Meryl J. Musicante, MD; David E. Cohen, MD, MPH</byline> <bylineText>Meryl J. Musicante, MD; David E. Cohen, MD, MPH; Emily C. Milam, MD </bylineText> <bylineFull>Meryl J. Musicante, MD; David E. Cohen, MD, MPH</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>35-42</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Approximately 20% of the general population has a contact allergy.1 Allergic contact dermatitis (ACD) is a delayed type IV hypersensitivity reaction mediated by</metaDescription> <articlePDF>299910</articlePDF> <teaserImage/> <title>Axillary Contact Dermatitis: An Update on Potential Allergens and Management</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2165</CMSID> </CMSIDs> <keywords> <keyword>contact dermatitis</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Audio | 2165</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">199</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026a1.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Axillary Contact Dermatitis: An Update on Potential Allergens and Management</title> <deck/> </itemMeta> <itemContent> <p class="abstract">The differential diagnosis of dermatoses in the axillae is broad. Contact dermatitis—both irritant and allergic—represents common etiologies. Axillary contact dermatitis can develop following exposure to a variety of irritants and/or allergens. Frequently implicated sources include deodorants, antiperspirants, detergents, soaps, and clothing. Fragrance, a ubiquitous ingredient within these products, as well as metals and dyes, are common causes of contact dermatitis. Clinical assessment, bedside diagnostic techniques, histopathology, and patch testing can aid in the diagnosis and help inform management directions.</p> <p> <em><em>Cutis.</em> 2024;113:35-42.</em> </p> <p>Approximately 20% of the general population has a contact allergy.<sup>1</sup> Allergic contact dermatitis (ACD) is a delayed type IV hypersensitivity reaction mediated by T lymphocytes.<sup>2</sup> Axillary ACD presentation is variable but typically includes an eczematous eruption with erythematous scaly patches or plaques. Common products in contact with the axillae include deodorants, antiperspirants, razors, bodywash, and clothing. </p> <p>Axillary skin is distinct from skin elsewhere on the body due to both anatomical characteristics and unique human self-care practices. Axillary skin has reduced barrier function, faster stratum corneum turnover, and altered lipid levels.<sup>3-5</sup> Moreover, the axillae often are subject to shaving or other hair removal practices that alter the local environment, as layers of stratum corneum and hair are mechanically removed, which causes irritation and predisposes the skin to enhanced sensitivity to topical exposures.<sup>6,7</sup> With the abundance of apocrine and eccrine glands, the axillae are prone to sweat, which also can accentuate contact allergy.<sup>2,3</sup> Other factors, such as occlusion and friction, contribute to axillary contact allergy.<sup>8,9<br/><br/></sup>Patch testing is the gold standard for the diagnosis of ACD and aids in identification of culprit allergens. A thorough patient history and examination of the rash distribution may provide further clues; for example, dermatitis due to a deodorant typically affects the vault, whereas textile dye dermatitis tends to spare the vault.<sup>10,11</sup> Baseline-limited patch testing detects up to two-thirds of clinically relevant allergens.<sup>12</sup> Therefore, patients may require subsequent testing with supplemental allergens. <br/><br/>The differential diagnosis for axillary lesions is broad—including inflammatory diseases such as irritant contact dermatitis and hidradenitis suppurativa, genetic disorders such as Hailey-Hailey disease, and infectious causes such as erythrasma—but may be narrowed with a thorough physical examination and patient history, histopathology, bedside diagnostic techniques (eg, scrapings and Wood lamp examination), and patch testing. Systemic contact dermatitis (SCD) or symmetrical drug-related intertriginous and flexural exanthema (SDRIFE) also may be suspected in cases of intertriginous dermatoses. <br/><br/>We review the potential allergens in products used on the axillae as well as the management of axillary ACD. We also discuss axillary dermatitis as a manifestation of SCD and SDRIFE. </p> <h3>Top Allergens in Products Used on the Axillae </h3> <p><i>Fragrance</i>—A 1982 North American Contact Dermatitis Group study on cosmetic products identified fragrances as the most common cause of ACD,<sup>13</sup> and this trend continues to hold true with more recent data.<sup>14</sup> The incidence of fragrance allergy may be increasing, with positive patch tests to a fragrance chemical in 10% of patch test clinic populations.<sup>15</sup> Fragrances are a ubiquitous ingredient in deodorants and antiperspirants, which are important sources implicated in the development and elicitation of fragrance ACD.<sup>16</sup> One study found that fragrance was present in 97 of 107 (90%) deodorants available at Walgreens pharmacies.<sup>11</sup></p> <p>In a study of patients with a history of an axillary rash caused by a deodorant spray, Johansen et al<sup>17</sup> reported that the likelihood of fragrance allergy is increased by a factor of 2.4. This risk of developing a fragrance allergy may be exacerbated in those who shave; Edman<sup>18</sup> reported that the odds ratio of developing a fragrance allergy among men who shave their beards was 2.9. Although there are no specific data on the effects of shaving on ACD, shaving in general can induce localized irritation and increase percutaneous absorption.<sup>19</sup> <br/><br/>The individual fragrance components in deodorants most likely to cause ACD include hydroxycitronellal, eugenol, and geraniol—all constituent ingredients in patch test formulations of fragrance mixture I.<sup>11,20</sup> Other common fragrance allergens associated with ACD include hydroxymethylpentylcyclohexenecarboxaldehyde, farnesol, and balsam of Peru.<sup>21-27</sup> Hydroperoxides of limonene and linalool, common fragrances in detergents and personal care products, are increasingly recognized as contact allergens and have been reported to cause axillary ACD from deodorants.<sup>28-30<br/><br/></sup>Dermatitis involving the bilateral axillary vaults wherever deodorant or antiperspirant was directly applied is the most common presentation of ACD due to fragrance (Figure 1).<sup>17</sup> An eczematous eruption is common, though scale may be less apparent than in nonflexural regions. Axillary ACD secondary to fragrances also may result from use of fragranced laundry detergents, fabric softeners, soaps, and perfumes, and may spare the vaults.<sup>10,29,31,32</sup> Less common presentations of axillary ACD due to fragrance include pigmented dermatoses; for example, ACD from an antiperspirant containing hydroperoxide of limonene presented as hyperpigmented patches with minimal erythema and scaling in the edges of the axillary folds.<sup>33,34<br/><br/></sup>Diagnosis of a fragrance ACD typically is made with a standard patch test series including fragrance mixture I and balsam of Peru, which may detect 75% and 50% of fragrance sensitivities, respectively.<sup>35</sup> Patch testing may be followed with a repeated open application test of the product in question.<sup>36</sup> Additionally, it may be appropriate to test for other fragrance allergens including balsam of Tolu, fragrance mixture II, lichen acid mix, and hydroxyperoxides of linalool and limonene (among other botanicals) if standard patch testing is negative and suspicion of fragrance ACD remains elevated.<sup>11</sup> <br/><br/><i>Propylene Glycol</i>—Propylene glycol (PG)—a versatile substance that functions as a solvent, humectant, emulsifier, stabilizer, and antimicrobial—is the second most common contact allergen present in deodorants.<sup>11</sup> It is prevalent in both personal care and household products, including deodorants, cosmetics, foods, toothpaste, cleaning agents, and detergents.<sup>11,37</sup> Propylene glycol is both an allergen and an irritant. Among deodorants/antiperspirants, PG is both a common irritant and allergen, as its concentration may be particularly high (as much as 73%).<sup>38</sup> One commonly reported example of PG contact dermatitis is from the topical medicament minoxidil.<sup>39,40<br/><br/></sup>Patch testing data have demonstrated a positivity rate for PG ranging between 0.1% to 3.8%. The variability in these findings likely is due to differences in the tested concentrations of PG, as higher concentrations sometimes required to elicit an allergic reaction also may create a stronger irritation effect.<sup>41</sup> Propylene glycol irritancy and the occlusive nature of the axillae may enhance sensitization to other allergens, as demonstrated by Agren-Jonsson and Magnusson,<sup>42</sup> who reported sensitization to propantheline bromide and trichlorocarbanilide in patients who used a lotion with 90% PG. Many PG-containing products beyond deodorants/antiperspirants may be applied to the axillae, including steroid creams, lotions, shaving creams, and bodywashes.<sup>38,43<br/><br/></sup>The diagnosis of PG allergy via patch testing is challenging and at times controversial given its irritant nature. False-positive irritant reactions have been documented, characterized by a weak reaction at 48 hours that is absent by 96 hours (decrescendo reaction). A reaction may not appear until 96 hours (crescendo reaction), which typically indicates a true contact allergy but in the case of PG also may be the substance acting as a “late irritant.”<sup>44</sup> Fast (<span class="body">&lt;</span>24 hours) and well-demarcated reactions suggest irritation.<sup>45</sup> Regardless, reactions to PG on patch testing, even those regarded as weak, may be considered relevant in consideration of the clinical context.<sup>37<br/><br/></sup><i>Aluminum</i>—Aluminum is the active ingredient in most antiperspirants, typically in the form of aluminum chloride, aluminum chlorohydrate, aluminum zirconium trichlorohydrex gly, or aluminum zirconium tetrachlorohydrex gly.<sup>46</sup> Aluminum mechanically obstructs the eccrine glands to reduce sweat.<sup>47</sup> Although aluminum is an uncommon allergen, a possible presentation of aluminum allergy is axillary vault dermatitis secondary to antiperspirant use.<sup>46</sup> Another potential manifestation is a ringlike reaction to the Finn Chambers (SmartPractice) used in patch testing.<sup>46</sup> In one case of aluminum-induced axillary dermatitis, a 28-year-old woman presented with eczema of the axillae, and subsequent patch testing revealed an allergy to aluminum chloride. The rash resolved upon cessation of use of an aluminum-containing deodorant.<sup>48<br/><br/></sup>Aluminum has been reported to cause granulomatous dermatitis in the axillae. This reaction typically presents as red-brown, pruritic papules limited to the area in which deodorant was applied, with histopathology revealing epithelioid granulomas.<sup>49-51</sup> <br/><br/>Alum deodorants—considered a natural alternative—contain aluminum bound to potassium or ammonium in the form of a crystal or powder. Alum crystal deodorants have been reported to cause both a typical erythematous pruritic dermatitis as well as a granulomatous dermatitis with red-brown papules.<sup>52,53</sup> The granulomatous dermatitis caused by either form of aluminum resolves with avoidance and use of topical steroids or topical tacrolimus.<sup>49,50,52,53<br/><br/></sup>The diagnosis of aluminum ACD via patch testing may be identified with empty Finn Chambers, which are metallic aluminum, or with patch placement of aluminum chloride hexahydrate, though the former is only positive in patients with a strong allergy.<sup>54,55</sup> In 2022, aluminum was named Allergen of the Year by the American Contact Dermatitis Society, with recommendations to conduct patch testing with aluminum chloride hexahydrate 10% rather than the traditional 2% to increase diagnostic yield.<sup>55</sup> Additionally, it is recommended that aluminum be included in baseline patch testing for children due to the high prevalence of aluminum allergy in children and early exposure via childhood vaccines.<sup>54-56</sup> In patients with aluminum allergy, providers may suggest purchasing aluminum-free deodorants or provide recipes for homemade deodorant that includes ingredients such as arrowroot powder, cornstarch, and diatomaceous earth.<sup>46<br/><br/></sup><i>Nickel</i>—Nickel is the most commonly identified contact allergen on patch testing yet an infrequent cause of axillary dermatitis. A case report from 2014 described axillary dermatitis in a woman that worsened during a positive patch test to nickel. Improvement was noted when the patient switched to titanium shaving razors.<sup>57</sup> Nickel allergy also may present in the form of SCD. In one report, a woman developed dermatitis of the flexural areas, including the axillae, 3 months after undergoing a sterilization procedure in which nickel-containing tubal implants were placed.<sup>58</sup> Patch testing revealed a positive reaction to nickel. The patient experienced complete resolution of the steroid-resistant dermatitis following removal of the implants via salpingectomy.<sup>58</sup> <br/><br/><i>Textile Dye</i>—In contrast to dermatitis caused by deodorants/antiperspirants, contact allergy to textile dyes presents as dermatitis involving the axillary borders but sparing the axillary vaults (Figures 2 and 3).<sup>10</sup> Other potential presentations of textile dye dermatitis include erythema multiforme–like eruptions and erythematous wheal–type reactions.<sup>59</sup> Textile dyes are classified as disperse vs nondisperse, with the majority of contact dermatoses caused by disperse dyes, specifically Disperse Orange 1, blue 106, and blue 124.<sup>60-62</sup> Ryberg et al<sup>61</sup> found that the axilla is one of the more common locations to be affected by textile dye allergy, particularly in women, which was further supported by Seidenari et al,<sup>63</sup><i> </i>who found that skin folds were affected in 27% of study participants allergic to textile dyes (N<span class="body">=</span>437), a finding that is likely due to friction, sweat, and occlusion.<sup>62</sup> In one case report of a patient with dermatitis caused by reactive dyes, the garment required 3 washes before the patient experienced resolution of dermatitis.<sup>64</sup> For patients with textile dye dermatitis, mitigation strategies include washing clothing before wearing, especially for darkly dyed items; avoiding tight clothing; wearing garments made of cotton, wool, silk, or linen; and choosing light-colored garments.<sup>9,64,65</sup></p> <h3>Axillary Dermatitis as a Manifestation of SCD and SDRIFE</h3> <p>Systemic contact dermatitis occurs when an individual who was previously sensitized to a particular allergen develops ACD of the skin with systemic exposure to that allergen or immunochemically related allergens. Exposure may occur via ingestion, inhalation, intravenous, intramuscular, and transepidermal routes.<sup>66</sup> Systemic contact dermatitis manifests in a variety of ways, including focal flares at sites of prior contact dermatitis (recall reaction), vesicular hand dermatitis, intertriginous eruptions including axillary dermatitis, and generalized eruptions.<sup>67</sup> </p> <p>Systemic contact dermatitis rarely involves systemic symptoms, and onset typically is within days of exposure. The 3 most common groups of allergens causing SCD are metals, medications, and plants and herbals.<sup>68</sup> These allergens have all been reported to cause axillary dermatitis via SCD.<sup>58,69,70</sup> Foods containing balsam of Peru that may lead to SCD include citrus, chocolate, tomato, and certain alcohols.<sup>70,71</sup> Patients with a positive patch test to balsam of Peru may experience improvement of their dermatitis after reduction of balsam of Peru–rich foods from their diet.<sup>70</sup> Metals implicated in SCD include mercury, nickel, and gold.<sup>72-74</sup> Finally, PG ingestion also has been implicated in cases of SCD.<sup>37</sup> <br/><br/>Symmetrical drug-related intertriginous and flexural exanthema is another condition that presents as intertriginous dermatitis and differs from SCD in that the eruption does not require presensitization; there may be no known prior exposure to the agent causing dermatitis. Historically, SDRIFE was described as baboon syndrome because of its frequent involvement of the buttocks with diffuse, well-demarcated, erythematous dermatitis resembling that of a baboon. This term is no longer used due to its insensitive nature and incomplete depiction of SDRIFE, which can affect body sites other than the buttocks.<sup>68,75,76</sup> Specific criteria to make this diagnosis include sharply demarcated and/or <i>V</i>-shaped erythema of the gluteal/perianal area, involvement of at least 1 other intertriginous or flexural region, symmetry of affected areas, and an absence of systemic symptoms.<sup>76</sup> There also may be papules, pustules, and vesicles present in affected areas. Symmetrical drug-related intertriginous and flexural exanthema most often is caused by <span class="body">β</span>-lactam antibiotics, but other associated drugs include chemotherapeutic agents, such as mitomycin C.<sup>76</sup> <br/><br/>Histopathology of both SCD and SDRIFE is variable and typically nonspecific, often revealing epidermal spongiosis and a perivascular mononuclear cell infiltrate with occasional neutrophils and eosinophils.<sup>76</sup> A case of SCD to mercury presenting as intertriginous dermatitis demonstrated a leukocytoclastic vasculitis pattern on biopsy.<sup>77</sup> <br/><br/>Systemic contact dermatitis is diagnosed via a patch test, while SDRIFE typically has a negative patch test result and requires oral rechallenge testing, which reproduces the rash within hours.<sup>78,79</sup> </p> <h3>Additional Allergens Causing Axillary ACD </h3> <p>Although fragrance is the most common allergen in deodorants, other ingredients have been shown to cause axillary ACD (Table).<sup>80-90</sup> In addition to these ingredients, allergens not previously mentioned that may be present in deodorants include lanolin, essential oils, and parabens.<sup>11</sup> Methylisothiazolinone in laundry detergent also has been found to instigate ACD.<sup>91</sup> Fragrances and preservatives in laundry detergents also may contribute to dermatitis.<sup>92</sup></p> <p>Other products that have caused axillary contact dermatitis include topical exposure to medicaments including clindamycin,<sup>93</sup> ethylenediamine in nystatin cream,<sup>94</sup> methylprednisolone acetate<sup>95</sup> and dipropylene glycol in a hydrocortisone lotion,<sup>96</sup> wood dusts from tropical hardwoods,<sup>97</sup> and tobacco.<sup>98</sup></p> <h3>Management of ACD</h3> <p>The most effective strategy in the management of patients with contact dermatitis is avoidance of the offending agent. Additionally, clinicians may recommend the use of topical steroids and/or calcineurin inhibitors to hasten resolution.<sup>2</sup> </p> <p>For patients with contact dermatitis, a clinician may recommend product substitutions with few potential allergens to use prior to patch testing. Patients with a fragrance allergy should look for products specifically labeled as “fragrance free” rather than “hypoallergenic” or “unscented,” as the latter two may still contain minimal amounts of fragrance.<sup>35</sup> Patients should be educated on the functions of the allergens to which they are allergic so they may adequately avoid potential sources of contact.<sup>99</sup> For suspected textile dye dermatitis, instructing patients to wash clothing before wearing and to avoid synthetic fabrics, dark dyes, and tightly fitted clothing may help.<sup>9,64,65</sup></p> <h3>Differential Diagnosis</h3> <p>The differential diagnosis for axillary lesions is broad, including infectious, inflammatory, and autoimmune etiologies. Irritant contact dermatitis (ICD) presents similar to ACD, though it is more immediate in onsetand typically demonstrates symptoms of burning and stinging rather than pruritus. Although histopathology is not reliable in differentiating ICD and ACD, it has been shown that focal parakeratosis is associated with ACD, whereas necrotic epidermal keratinocytes are found in ICD.<sup>100</sup> </p> <p>Intertrigo presents as large, erythematous, opposing patches or plaques confined to inguinal, submammary, axillary, and/or abdominal folds. Findings of beefy red erythema and peripheral satellite pustules may implicate presence of <i>Candida</i>, which can be identified with potassium hydroxide preparations. <br/><br/>Inverse psoriasis presents as sharply demarcated, erythematous, moist, smooth plaques or patches with minimal scale. The most common area of involvement is the inguinal folds, followed by the axillae, inframammary folds, perianal area, umbilicus, and retroauricular areas. Involvement of the elbows and knees or a positive family history of psoriasis may be useful knowledge in establishing the diagnosis. A biopsy may show dermal eosinophils, epidermal spongiosis, and focal serum in the scale, in addition to features of typical psoriasis plaques.<sup>101</sup> <br/><br/>Seborrheic dermatitis typically is an erythematous eruption, often with yellowish greasy scale. Simultaneous involvement of the face and scalp may be noted. Although typically a clinical diagnosis, biopsy demonstrates shoulder parakeratosis with follicular plugging and lymphocytic exocytosis.<br/><br/>Hailey-Hailey disease (also called benign familial pemphigus) is an autosomal-dominant genetic condition presenting as moist, malodorous, painful, vegetative plaques, patches, or scaly pustules in flexural areas, frequently with flaccid blisters. Lesions are provoked by traumatic stimuli. Onset occurs in the second to fourth decades and may improve with age. The diagnosis is confirmed by biopsy, which demonstrates acantholysis of the epidermis. The moist superficial patches of Hailey-Hailey disease help distinguish it from comparably drier Darier disease, the other acantholytic disease of the axillae.<br/><br/>Granular parakeratosis (also called hyperkeratotic flexural erythema) is an uncommon dermatosis most often observed in middle-aged women. It presents as red-brown keratotic papules coalescing into plaques, often with overlying scale in intertriginous areas. This disorder may be related to exposure to aluminum, a key component of antiperspirants.<sup>102</sup> Diagnosis with a skin biopsy demonstrates granular parakeratosis. <br/><br/>Infections most commonly include erythrasma, tinea, and candidiasis. Erythrasma caused by <i>Corynebacterium minutissimum</i> may present in the axillae and/or groin with sharply demarcated, red-brown patches. Wood lamp examination reveals coral red fluorescence. Tinea corporis, a dermatophyte infection, may present as scaly erythematous plaques with advancing borders and central clearing. Fungal cultures and potassium hydroxide preparations are useful to confirm the diagnosis.<br/><br/>Pseudofolliculitis barbae most often is thought of as a condition affecting the beard in Black men, but it also may present in individuals of all races who shave the axillary and inguinal regions. Typical features include pruritic inflammatory papules and pustules with surrounding erythema and hyperpigmentation.<br/><br/>Fox-Fordyce disease is a disorder of the apocrine sweat glands that presents as several flesh-colored, perifollicular, monomorphic papules in the axillae. It typically is a disease of young females and also can involve the areola and vulva. Histopathology may show hyperkeratosis, irregular acanthosis, and dilated sweat glands. <br/><br/>Hidradenitis suppurativa is a chronic inflammatory condition that presents with multiple cysts; nodules; abscesses; sinus tract formation; and suppuration of the axillary, anogenital, and sometimes inframammary areas, typically at the onset of puberty. The diagnosis is best supported by history and physical examination, which may be notable for recurrent abscesses, draining tracts, double comedones, and ropelike scarring.<br/><br/>Extramammary Paget disease is a rare malignancy affecting apocrine gland–bearing areas, including axillary and genital regions. It most commonly presents as a unilateral or asymmetric, scaly, erythematous plaque. Histopathology demonstrates Paget cells with abundant clear cytoplasm and pleomorphic nuclei, typically grouped in the lower portion of the epidermis.</p> <h3>Final Thoughts</h3> <p>Axillary dermatoses often can be challenging to diagnose given the range of pathologies that can present in intertriginous areas. Allergic contact dermatitis is a common culprit due to unique anatomical considerations and self-care practices, including shaving/hair removal; use of deodorants, antiperspirants, bodywashes, and clothing; and frictional and moisture influences. The most likely offender among contact allergens is fragrance, but other possibilities to consider include PG, preservatives, aluminum, nickel, and textile dyes. Albeit less common, systemic exposure to allergens may result in SCD and SDRIFE with a rash in intertriginous zones, including the axillae. Additionally, other infectious, inflammatory, and autoimmune etiologies should be considered and ruled out.</p> <p>Patch testing is the most reliable method to diagnose suspected ACD. Once confirmed, management includes the use of topical steroids and avoidance of the causative agent. Additionally, patients should be informed of the American Contact Dermatitis Society Contact Allergen Management Program (<a href="https://www.contactderm.org/patient-support/camp-access">https://www.contactderm.org/patient-support/camp-access</a>), which provides patients with useful information on products that are safe to use based on their patch testing results.</p> <h2>References</h2> <p class="reference"> 1. Alinaghi F, Bennike NH, Egeberg A, et al. Prevalence of contact allergy in the general population: a systematic review and meta-analysis. <i>Contact Dermatitis. </i>2019;80:77-85.</p> <p class="reference"> 2. Brar KK. A review of contact dermatitis. <i>Ann Allergy Asthma Immunol. </i>2021;126:32-39.<br/><br/> 3. Evans RL, Marriott RE, Harker M. Axillary skin: biology and care. <i>Int J Cosmet Sci. </i>2012;34:389-395.<br/><br/> 4. Watkinson A, Lee RS, Moore AE, et al. Is the axilla a distinct skin phenotype? <i>Int J Cosmet Sci. </i>2007;29:60.<br/><br/> 5. Wu JQ, Kilpatrick-Liverman L. Characterizing the composition of underarm and forearm skin using confocal raman spectroscopy. <i>Int J Cosmet Sci. </i>2011;33:257-262.<br/><br/> 6. Marti VP, Lee RS, Moore AE, et al. Effect of shaving on axillary stratum corneum. <i>Int J Cosmet Sci. </i>2003;25:193-198.<br/><br/> 7. Turner GA, Moore AE, Marti VPJ, et al. Impact of shaving and anti-perspirant use on the axillary vault. <i>Int J Cosmet Sci. </i>2007;29:31-38.<br/><br/> 8. Zhai H, Maibach HI. Skin occlusion and irritant and allergic contact dermatitis: an overview. <i>Contact Dermatitis. </i>2001;44:201-206.<br/><br/> 9. Lazarov A. Textile dermatitis in patients with contact sensitization in Israel: a 4-year prospective study. <i>J Eur Acad Dermatol Venereol. </i>2004;18:531-537.<br/><br/> 10. Nelson JL, Mowad CM. Allergic contact dermatitis: patch testing beyond the TRUE Test. <i>J Clin Aesthet Dermatol. </i>2010;3:36-41.<br/><br/> 11. Zirwas MJ, Moennich J. Antiperspirant and deodorant allergy: diagnosis and management. <i>J Clin Aesthet Dermatol. </i>2008;1:38-43.<br/><br/> 12. DeKoven JG, Warshaw EM, Reeder MJ, et al. North American Contact Dermatitis Group Patch Test Results: 2019-2020. <i>Dermatitis. </i>2023;34:90-104.<br/><br/> 13. Eiermann HJ, Larsen W, Maibach HI, et al. Prospective study of cosmetic reactions: 1977-1980. North American Contact Dermatitis Group. <i>J Am Acad Dermatol. </i>1982;6:909-917.</p> <p class="reference"> 14. González-Muñoz P, Conde-Salazar L, Vañó-Galván S. Allergic contact dermatitis caused by cosmetic products. <i>Actas Dermosifiliogr. </i>2014;105:822-832.<br/><br/> 15. Gerberick GF, Robinson MK, Felter SP, et al. Understanding fragrance allergy using an exposure-based risk assessment approach. <i>Contact Dermatitis. </i>2001;45:333-340.</p> <p class="reference"> 16. Heisterberg MV, Menne T, Andersen KE, et al. Deodorants are the leading cause of allergic contact dermatitis to fragrance ingredients. <i>Contact Dermatitis. </i>2011;64:258-264.<br/><br/> 17. Johansen JD, Andersen TF, Kjoller M, et al. Identification of risk products for fragrance contact allergy: a case-referent study based on patients’ histories. <i>Am J Contact Dermat. </i>1998;9:80-86.<br/><br/> 18. Edman B. The influence of shaving method on perfume allergy. <i>Contact Dermatitis. </i>1994;31:291-292.<br/><br/> 19. Hamza M, Tohid H, Maibach H. Shaving effects on percutaneous penetration: clinical implications. <i>Cutan Ocul Toxicol. </i>2015;34:335-343.<br/><br/> 20. Geier J, Uter W, Lessmann H, et al. Fragrance mix I and II: results of breakdown tests. <i>Flavour Fragr J. </i>2015;30:264-274.<br/><br/> 21. Handley J, Burrows D. Allergic contact dermatitis from the synthetic fragrances Lyral and acetyl cedrene in separate underarm deodorant preparations. <i>Contact Dermatitis. </i>1994;31:288-290.<br/><br/> 22. Hendriks SA, Bousema MT, van Ginkel CJ. Allergic contact dermatitis from the fragrance ingredient Lyral in underarm deodorant. <i>Contact Dermatitis. </i>1999;41:119.<br/><br/> 23. Jacob SE. Allergic contact dermatitis from lyral in an aerosol deodorant. <i>Dermatitis. </i>2008;19:216-217.<br/><br/> 24. Gilpin S, Maibach H. Allergic contact dermatitis caused by farnesol: clinical relevance. <i>Cutan Ocul Toxicol. </i>2010;29:278-287.<br/><br/> 25. Goossens A, Merckx L. Allergic contact dermatitis from farnesol in a deodorant. <i>Contact Dermatitis. </i>1997;37:179-180.<br/><br/> 26. Schnuch A, Uter W, Geier J, et al. Contact allergy to farnesol in 2021 consecutively patch tested patients. Results of the IVDK. <i>Contact Dermatitis. </i>2004;50:117-121.<br/><br/> 27. Uter W, Geier J, Schnuch A, et al. Patch test results with patients’ own perfumes, deodorants and shaving lotions: results of the IVDK 1998–2002. <i>J Eur Acad Dermatol Venereol. </i>2007;21:374-379.<br/><br/> 28. Dittmar D, Schuttelaar MLA. Contact sensitization to hydroperoxides of limonene and linalool: results of consecutive patch testing and clinical relevance. <i>Contact Dermatitis. </i>2019;80:101-109.<br/><br/> 29. Yazar K, Johnsson S, Lind M-L, et al. Preservatives and fragrances in selected consumer-available cosmetics and detergents. <i>Contact Dermatitis. </i>2011;64:265-272.<br/><br/> 30. Isaksson M, Karlberg A-T, Nilsson U. Allergic contact dermatitis caused by oxidized linalool in a deodorant. <i>Contact Dermatitis. </i>2019;81:213-214.<br/><br/> 31. Chen J, Yi Z, Sun R, et al. Analysis of fragrance allergens in personal care products, toys, and water samples: a review. <i>J AOAC Int. </i>2022;105:396-412.<br/><br/> 32. Larsen WG. Perfume dermatitis. <i>J Am Acad Dermatol. </i>1985;12:1-9.<br/><br/> 33. Pincelli C, Magni R, Motolese A. Pigmented contact dermatitis from deodorant. <i>Contact Dermatitis. </i>1993;28:305-306.<br/><br/> 34. Kwong HL, Lim SPR. Pigmented contact dermatitis in the axillae caused by hydroperoxides of limonene. <i>JAAD Case Reports. </i>2020;6:476-478.<br/><br/> 35. Marks J, Anderson B, DeLeo V. <i>Contact and Occupational Dermatology. </i>4th ed. Jaypee; 2016.<br/><br/> 36. Johansen JD. Fragrance contact allergy: a clinical review. <i>Am J Clin Dermatol. </i>2003;4:789-798.<br/><br/> 37. McGowan MA, Scheman A, Jacob SE. Propylene glycol in contact dermatitis: a systematic review. <i>Dermatitis. </i>2018;29:6-12.<br/><br/> 38. Fiume MM, Bergfeld WF, Belsito DV, et al. Safety assessment of propylene glycol, tripropylene glycol, and PPGs as used in cosmetics. <i>Int J Toxicol. </i>2012;31(5 suppl):245S-260S.<br/><br/> 39. Farrar CW, Bell HK, King CM. Allergic contact dermatitis from propylene glycol in Efudix cream. <i>Contact Dermatitis. </i>2003;48:345.<br/><br/> 40. Friedman ES, Friedman PM, Cohen DE, et al. Allergic contact dermatitis to topical minoxidil solution: etiology and treatment. <i>J Am Acad Dermatol. </i>2002;46:309-312.<br/><br/> 41. Lessmann H, Schnuch A, Geier J, et al. Skin-sensitizing and irritant properties of propylene glycol. <i>Contact Dermatitis. </i>2005;53:247-259.<br/><br/> 42. Agren-Jonsson S, Magnusson B. Sensitization to propantheline bromide, trichlorocarbanilide and propylene glycol in an antiperspirant. <i>Contact Dermatitis. </i>1976;2:79-80.<br/><br/> 43. Catanzaro JM, Smith JG Jr. Propylene glycol dermatitis. <i>J Am Acad Dermatol. </i>1991;24:90-95.<br/><br/> 44. Jacob SE, Scheman A, McGowan MA. Propylene glycol. <i>Dermatitis. </i>2018;29:3-5.<br/><br/> 45. Carlson S, Gipson K, Nedorost S. Relevance of doubtful (“equivocal”) late patch-test readings. <i>Dermatitis. </i>2010;21:102-108.<br/><br/> 46. Kullberg SA, Ward JM, Liou YL, et al. Cutaneous reactions to aluminum. <i>Dermatitis. </i>2020;31:335-349.<br/><br/> 47. Benohanian A. Antiperspirants and deodorants. <i>Clin Dermatol. </i>2001;19:398-405.</p> <p class="reference"> 48. Garg S, Loghdey S, Gawkrodger DJ. Allergic contact dermatitis from aluminum in deodorants. <i>Contact Dermatitis. </i>2010;62:57-58.<br/><br/> 49. Montemarano AD, Sau P, Johnson FB, et al. Cutaneous granulomas caused by an aluminum-zirconium complex: an ingredient of antiperspirants. <i>J Am Acad Dermatol. </i>1997;37:496-498.<br/><br/> 50. Rubin L, Slepyan AH, Weber LF, et al. Granulomas of the axillae caused by deodorants. <i>JAMA. </i>1956;162:953-955.<br/><br/> 51. Williams S, Freemont AJ. Aerosol antiperspirants and axillary granulomata. <i>Br Med J (Clin Res Ed). </i>1984;288:1651-1652.<br/><br/> 52. Gallego H, Lewis EJ, Crutchfield CE 3rd. Crystal deodorant dermatitis: irritant dermatitis to alum-containing deodorant. <i>Cutis. </i>1999;64:65-66.<br/><br/> 53. Leventhal JS, Farhadian JA, Miller KE, et al. Crystal deodorant-induced axillary granulomatous dermatitis. <i>Int J Dermatol. </i>2014;53:e59-e60.<br/><br/> 54. Siemund I, Dahlin J, Hindsén M, et al. Contact allergy to two aluminum salts in consecutively patch-tested dermatitis patients. <i>Dermatitis. </i>2022;3:31-35.<br/><br/> 55. Bruze M, Netterlid E, Siemund I. Aluminum-allergen of the year 2022. <i>Dermatitis. </i>2022;33:10-15.<br/><br/> 56. Goiset A, Darrigade A-S, Labrèze C, et al. Aluminum sensitization in a French paediatric patch test population. <i>Contact Dermatitis. </i>2018;79:382-383.<br/><br/> 57. Admani S, Matiz C, Jacob SE. Nickel allergy—a potential cause of razor dermatitis. <i>Pediatr Dermatol. </i>2014;31:392-393.<br/><br/> 58. Bibas N, Lassere J, Paul C, et al. Nickel-induced systemic contact dermatitis and intratubal implants: the baboon syndrome revisited. <i>Dermatitis. </i>2013;24:35-36.<br/><br/> 59. Seidenari S, Manzini BM, Ddanese P. Contact sensitization to textile dyes: description of 100 subjects. <i>Contact Dermatitis. </i>1991;24:253-258.<br/><br/> 60. Hatch KL, Maibach HI. Textile dye allergic contact dermatitis prevalence. <i>Contact Dermatitis. </i>2000;42:187-195.<br/><br/> 61. Ryberg K, Isaksson M, Gruvberger B, et al. Contact allergy to textile dyes in southern Sweden. <i>Contact Dermatitis. </i>2006;54:313-321.<br/><br/> 62. Pratt M, Taraska V. Disperse blue dyes 106 and 124 are common causes of textile dermatitis and should serve as screening allergens for this condition. <i>Dermatitis. </i>2000;11:30-41.<br/><br/> 63. Seidenari S, Giusti F, Massone F, et al. Sensitization to disperse dyes in a patch test population over a five-year period. <i>Am J Contact Dermat. </i>2002;13:101-107.<br/><br/> 64. Moreau L, Goossens A. Allergic contact dermatitis associated with reactive dyes in a dark garment: a case report. <i>Contact Dermatitis. </i>2005;53:150-154.<br/><br/> 65. Svedman C, Engfeldt M, Malinauskiene L. Textile contact dermatitis: how fabrics can induce dermatitis. <i>Curr Treat Options Allergy. </i>2019;6:103-111.<br/><br/> 66. Jacob SE, Zapolanski T. Systemic contact dermatitis. <i>Dermatitis. </i>2008;19:9-15.<br/><br/> 67. Hindsén M, Bruze M, Christensen OB. Flare-up reactions after oral challenge with nickel in relation to challenge dose and intensity and time of previous patch test reactions. <i>J Am Acad Dermatol. </i>2001;44:616-623.<br/><br/> 68. Winnicki M, Shear NH. A systematic approach to systemic contact dermatitis and symmetric drug-related intertriginous and flexural exanthema (SDRIFE): a closer look at these conditions and an approach to intertriginous eruptions. <i>Am J Clin Dermatol. </i>2011;12:171-180.<br/><br/> 69. Kalita BJ, Das S, Dutta B. Itraconazole-induced symmetrical drug-related intertriginous and flexural exanthema (SDRIFE): a rare occurrence. <i>Int J Dermatol. </i>2020;59:e419-e421.<br/><br/> 70. Salam TN, Fowler JF Jr. Balsam-related systemic contact dermatitis. <i>J Am Acad Dermatol. </i>2001;45:377-381.<br/><br/> 71. Ramachandran V, Cline A, Summey B, et al. Systemic contact dermatitis related to alcoholic beverage consumption. <i>Dermatol Online J. </i>2019;25:13030/qt3zg853qv.<br/><br/> 72. Moreno-Ramírez D, García-Bravo B, Pichardo AR, et al. Baboon syndrome in childhood: easy to avoid, easy to diagnose, but the problem continues. <i>Pediatr Dermatol. </i>2004;21:250-253.<br/><br/> 73. Dou X, Liu L-L, Zhu X-J. Nickel-elicited systemic contact dermatitis. <i>Contact Dermatitis. </i>2003;48:126-129.<br/><br/> 74. Möller H, Ohlsson K, Linder C, et al. The flare-up reactions after systemic provocation in contact allergy to nickel and gold. <i>Contact Dermatitis. </i>1999;40:200-204.<br/><br/> 75. Andersen KE, Hjorth N, Menné T. The baboon syndrome: systemically-induced allergic contact dermatitis. <i>Contact Dermatitis. </i>1984;10:97-100.<br/><br/> 76. Häusermann P, Harr T, Bircher AJ. Baboon syndrome resulting from systemic drugs: is there strife between SDRIFE and allergic contact dermatitis syndrome? <i>Contact Dermatitis. </i>2004;51:297-310.<br/><br/> 77. Tan MG, Pratt MD, Burns BF, et al. Baboon syndrome from mercury showing leukocytoclastic vasculitis on biopsy. <i>Contact Dermatitis. </i>2020;83:415-417.<br/><br/> 78. Handisurya A, Stingl G, Wöhrl S. SDRIFE (baboon syndrome) induced by penicillin. <i>Clin Exp Dermatol. </i>2009;34:355-357.<br/><br/> 79. Akay BN, Sanli H. Symmetrical drug-related intertriginous and flexural exanthem due to oral risperidone. <i>Pediatr Dermatol. </i>2009;26:214-216.</p> <p class="reference"> 80. Amaro C, Santos R, Cardoso J. Contact allergy to methylisothiazolinone in a deodorant. <i>Contact Dermatitis. </i>2011;64:298-299.<br/><br/> 81. Goh CL. Dermatitis from chlorphenesin in a deodorant. <i>Contact Dermatitis. </i>1987;16:287.<br/><br/> 82. Taghipour K, Tatnall F, Orton D. Allergic axillary dermatitis due to hydrogenated castor oil in a deodorant. <i>Contact Dermatitis. </i>2008;58:168-169.<br/><br/> 83. Sheu M, Simpson EL, Law S V, et al. Allergic contact dermatitis from a natural deodorant: a report of 4 cases associated with lichen acid mix allergy. <i>J Am Acad Dermatol. </i>2006;55:332-337.<br/><br/> 84. Pastor-Nieto M-A, Gatica-Ortega M-E, Alcántara-Nicolás F-D-A, et al. Allergic contact dermatitis resulting from cetyl PEG/PPG-10/1 dimethicone in a deodorant cream. <i>Contact Dermatitis. </i>2018;78:236-239.<br/><br/> 85. Corazza M, Lombardi AR, Virgili A. Non-eczematous urticarioid allergic contact dermatitis due to Eumulgin L in a deodorant. <i>Contact Dermatitis. </i>1997;36:159-160.<br/><br/> 86. van Ketel WG. Allergic contact dermatitis from propellants in deodorant sprays in combination with allergy to ethyl chloride. <i>Contact Dermatitis. </i>1976;2:115-119.<br/><br/> 87. Shmunes E, Levy EJ. Quaternary ammonium compound contact dermatitis from a deodorant. <i>Arch Dermatol. </i>1972;105:91-93.<br/><br/> 88. Bruze M, Johansen JD, Andersen KE, et al. Deodorants: an experimental provocation study with cinnamic aldehyde. <i>J Am Acad Dermatol. </i>2003;48:194-200.<br/><br/> 89. Hann S, Hughes TM, Stone NM. Flexural allergic contact dermatitis to benzalkonium chloride in antiseptic bath oil. <i>Br J Dermatol. </i>2007;157:795-798.<br/><br/> 90. Aeling JL, Panagotacos PJ, Andreozzi RJ. Allergic contact dermatitis to vitamin E aerosol deodorant. <i>Arch Dermatol. </i>1973;108:579-580.<br/><br/> 91. Cotton CH, Duah CG, Matiz C. Allergic contact dermatitis due to methylisothiazolinone in a young girl’s laundry detergent. <i>Pediatr Dermatol. </i>2017;34:486-487.<br/><br/> 92. Magnano M, Silvani S, Vincenzi C, et al. Contact allergens and irritants in household washing and cleaning products. <i>Contact Dermatitis. </i>2009;61:337-341.<br/><br/> 93. Voller LM, Kullberg SA, Warshaw EM. Axillary allergic contact dermatitis to topical clindamycin. <i>Contact Dermatitis. </i>2020;82:313-314.<br/><br/> 94. Iammatteo M, Akenroye A, Jariwala S, et al. Severe contact dermatitis due to ethylenediamine dihydrochloride in nystatin cream. <i>J Allergy Clin Immunol Pract. </i>2017;5:1448-1450.<br/><br/> 95. Coskey RJ, Bryan HG. Contact dermatitis due to methylprednisolone. <i>JAMA. </i>1967;199:136.<br/><br/> 96. Peterson MY, Han J, Warshaw EM. Allergic contact dermatitis from dipropylene glycol in hydrocortisone lotion. <i>Contact Dermatitis. </i>2022;87:112-114.<br/><br/> 97. Ferreira O, Cruz MJ, Mota A, et al. Erythema multiforme-like lesions revealing allergic contact dermatitis to exotic woods. <i>Cutan Ocul Toxicol. </i>2012;31:61-63.<br/><br/> 98. Abraham NF, Feldman SR, Vallejos Q, et al. Contact dermatitis in tobacco farmworkers. <i>Contact Dermatitis. </i>2007;57:40-43.<br/><br/> 99. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. <i>J Am Acad Dermatol. </i>2016;74:1043-1054.<br/><br/>100. Frings VG, Böer-Auer A, Breuer K. Histomorphology and immunophenotype of eczematous skin lesions revisited-skinbiopsies are not reliable in differentiating allergic contact dermatitis, irritant contact dermatitis, and atopic dermatitis. <i>Am J Dermatopathol. </i>2018;40:7-16.<br/><br/>101. Knabel M, Mudaliar K. Histopathologic features of inverse psoriasis. <i>J Cutan Pathol. </i>2022;49:246-251.<br/><br/>102. Fujii M, Kishibe M, Honma M, et al. Aluminum chloride-induced apoptosis leads to keratinization arrest and granular parakeratosis. <i>Am J Dermatopathol. </i>2020;42:756-761.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. Musicante is from The University of Tennessee Health Science Center College of Medicine, Memphis. Drs. Cohen and Milam are from the Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York.</p> <p class="disclosure">Drs. Musicante and Milam report no conflict of interest. Dr. Cohen has been a consultant for and received honoraria from Cosmetic Ingredient Review; Ferndale Laboratories, Inc; FIDE; LEO Pharma; Medimetriks; Novartis (past); SFJ Pharmaceuticals, Inc (past); and UCB. He also owns stock or has stock options in Evommune, Kadmon (past), and Timber Pharmaceuticals, and is on the board of directors for Evommune, Kadmon (past), and Timber Pharmaceuticals.<br/><br/>Correspondence: Emily C. Milam, MD, 240 E 38th St, Floor 12, New York, NY 10016 (Emily.Milam@nyulangone.org).doi:10.12788/cutis.0930</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>The differential diagnosis of axillary dermatitis is broad. Contact dermatitis—both irritant and allergic—represents common etiologies.</li> <li>Understanding the clinical features and range of potential sources in axillary contact dermatitis allows for efficient recognition and elimination of causative exposure.</li> <li>For cases of suspected allergic contact dermatitis, patch testing and subsequent allergen avoidance are paramount in the management of axillary eruptions.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • The differential diagnosis of axillary dermatitis is broad. Contact dermatitis—both irritant and allergic—represents common etiologies.
  • Understanding the clinical features and range of potential sources in axillary contact dermatitis allows for efficient recognition and elimination of causative exposure.
  • For cases of suspected allergic contact dermatitis, patch testing and subsequent allergen avoidance are paramount in the management of axillary eruptions.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026A1.SIG
Disable zoom
Off

Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses

Article Type
Changed
Tue, 01/09/2024 - 12:48
Display Headline
Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses

As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.2 Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle.

Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.3 One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.

Discoid Lupus Erythematosus

Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,4 and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,5 India,6 and Japan7 have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,5 which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).5,8

Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.5 It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,9 as SCCs arising in DLE lesions virtually always display prominent solar elastosis6; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.10

Kuraitis_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Invasive%20squamous%20cell%20carcinoma%20arising%20within%20a%20labial%20discoid%20lupus%20erythematosus%20lesion.%20This%20patient%E2%80%99s%20lesions%20were%20present%20for%20approximately%206%20years%20prior%20to%20presentation%20for%20carcinoma.%3C%2Fp%3E

Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,11 thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.12 However, testing for HPV is not routinely performed in these cases.

Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips.

Lichen Planus

Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,13 variants of lichen planus may predispose patients to SCC.

 

 

Oral Lichen Planus—Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,14 compared to a reference rate of 0.2% for oral SCC.15 A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.16 The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.15 Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.17

Hypertrophic Lichen Planus—The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.13 A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.18

Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.19 In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.20

It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.21,22 In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment.

Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.23 Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.Nail Lichen Planus—Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.24 Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.25,26

Lichen Sclerosus

Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.27 It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N=78) may have adjacent LS.28

Kuraitis_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Poorly%20differentiated%20squamous%20cell%20carcinoma%20arising%20within%20vulvar%20lichen%20sclerosus.%20This%20patient%E2%80%99s%20dermatosis%20was%20present%20for%20approximately%207%20years%20prior%20to%20presentation%20for%20carcinoma.%3C%2Fp%3E

In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.29 Other studies have estimated a lag time of 4 years until SCC presentation.30 An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.31 It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.32 Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.33 Extragenital LS does not appear to have similar potential for malignant transformation.34

 

 

In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.35 Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.35 In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.36 These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.

The rate of SCC arising in male genital LS may approach 8.4%,37 with a lag time of 17 years from onset of LS to SCC diagnosis.38 Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.39 Male penile SCC in a background of LS may not necessarily be HPV associated.40

Marjolin Ulcer

Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.41 Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.42 Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.43 Cohort studies of MU on the lower extremities have observed lag times of 26.444 and 37.945 years, with both studies also noting relatively high rates of local recurrence.

The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,41 and local accumulation of toxin may promote mutagenesis.46 Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system47 and become even more aggressive as the tumor accumulates.48 Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.49

Hidradenitis Suppurativa

As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.50 Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.51 Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, β-defensins, and interleukins.52

A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.53 After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.53 A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.54 As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.55

Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),54,56 despite HS being more prevalent in females (2:1 ratio).57 Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women58; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.

 

 

Necrobiosis Lipoidica Diabeticorum

Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.59 The lag time was 21.5 years; 31% of cases (N=14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized.

Epidermolysis Bullosa

Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.60 Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.61 Although SCCs tend to be well differentiated in RDEB (73.9%),61 they also exhibit highly aggressive behavior.62 In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.63

As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.64 There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.65 Mutations in p53,66 local alterations in transforming growth factor β activity,67 and downstream matrix metalloproteinase activity68 have been implicated.

Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.69 Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.70 A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.71

There does not appear to be evidence for HPV involvement in RDEB-associated SCC.72 Squamous cell carcinoma development in RDEB appears to be multifactorial,73 but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.73

Conclusion

Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.


References
  1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121:2373-2380. doi:10.1002/ijc.23173
  2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436-444. doi:10.1038/nature07205
  3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2011;2:98. doi:10.3389/fimmu.2011.00098
  4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. Clin Dermatol. 2004;22:121-124. doi:10.1016/j.clindermatol.2003.12.018
  5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. J Am Acad Dermatol. 2012;66:695-696. doi:10.1016 /j.jaad.2011.09.033
  6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. Lupus. 2015;24:1562-1566. doi:10.1177/0961203315599245
  7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. Eur J Dermatol. 2016;26:395-396. doi:10.1684/ejd.2016.2780
  8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. J Clin Oncol. 2005;23:759-765. doi:10.1200/JCO.2005.02.155
  9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol. 2018;125:e22-e30. doi:10.1016/j.oooo.2017.08.012
  10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. J Clin Rheumatol. 2011;17:35-36. doi:10.1097/RHU.0b013e3182051928
  11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol. 2015;27:454-460. doi:10.1097/BOR.0000000000000202
  12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. Cutis. 2010;86:195-198.
  13. Sigurgeirsson B, Lindelöf B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. Arch Dermatol. 1991;127:1684-1688. doi:10.1001/archderm.127.11.1684
  14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc. 2014;145:45-56. doi:10.14219/jada.2013.10
  15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol. 2019;58:296-301. doi:10.1111/ijd.14215
  16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol. 2017;68:92-102. doi:10.1016/j.oraloncology.2017.03.012
  17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. Odontology. 2017;105:262-266. doi:10.1007/s10266-016-0255-4
  18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. Dermatol Surg. 2015;41:1411-1418. doi:10.1097/DSS.0000000000000565
  19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. Pediatr Dermatol. 2015;32:e98-e101. doi:10.1111/pde.12549
  20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. Dermatol Online J. 2003;9:10.
  21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. J Cutan Pathol. 2012;39:243-250. doi:10.1111/j.1600-0560.2011.01834.x
  22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. Dermatol Surg. 2018;44:1466-1470. doi:10.1097/DSS.0000000000001465
  23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. JAAD Case Rep. 2017;3:151-154. doi: 10.1016/j.jdcr.2017.01.020
  24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. J Am Acad Dermatol. 2005;53:908-909. doi:10.1016/j.jaad.2005.04.052
  25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. J Am Acad Dermatol. 2011;64:1147-1153. doi:10.1016/j.jaad.2010.02.057
  26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. J Am Acad Dermatol. 2019;81:1358-1370. doi:10.1016/j.jaad.2019.03.070
  27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416. doi:10.1016/0190-9622(95)90060-8
  28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. Br J Obstet Gynaecol. 1990;97:1135-1139. doi:10.1111/j.1471-0528.1990.tb02502.x
  29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:1224-1230. doi:10.1158/1055-9965.EPI-16-0019
  30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol. 1998;29:932-948. doi:10.1016/s0046-8177(98)90198-8
  31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. J Low Genit Tract Dis. 2016;20:180-183. doi:10.1097/LGT.0000000000000186
  32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. JAMA Dermatol. 2015;151:1059-1060. doi:10.1001/jamadermatol.2015.0644
  33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. Am J Surg Pathol. 2018;42:828-835. doi:10.1097/PAS.0000000000001047
  34. Val I, Almeida G. An overview of lichen sclerosus. Clin Obstet Gynecol. 2005;48:808-817. doi:10.1097/01.grf.0000179635.64663.3d
  35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. JAMA Dermatol. 2015;151:1061-1067. doi:10.1001/jamadermatol.2015.0643
  36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. Arch Dermatol. 2004;140:709-712. doi:10.1001/archderm.140.6.709
  37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. Urology. 2010;76(2 suppl 1):S24-S35. doi:10.1016/j.urology.2010.04.007
  38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911-914. doi:10.1016/s0190-9622(99)70245-8
  39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. BJU Int. 2013;111:970-976. doi:10.1111/j.1464-410X.2012.11773.x
  40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. Am J Surg Pathol. 2003;27:1448-1453. doi:10.1097/00000478-200311000-00007
  41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64. doi:10.1016/j.jcws.2012.04.001
  42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? Burns. 2005;31:421-431. doi:10.1016/j.burns.2005.02.008
  43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? Int Wound J. 2019;16:479-485. doi:10.1111/iwj.13058
  44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. Ann Plast Surg. 2019;83:518-522. doi:10.1097/SAP.0000000000001995
  45. Karasoy Yesilada A, Zeynep Sevim K, Özgur Sucu D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. J Cutan Med Surg. 2013;17:404-409. doi:10.2310/7750.2013.13016
  46. Bazalin´ski D, Przybek-Mita J, Baran´ska B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. Contemp Oncol (Pozn). 2017;21:197-202. doi:10.5114/wo.2017.70109
  47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. Ann Plast Surg. 1986;16:42-48. doi:10.1097/00000637-198601000-00004
  48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? Plast Reconstr Surg. 1976;57:66-69.
  49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. Plast Reconstr Surg. 2009;123:184-191. doi:10.1097/PRS.0b013e3181904d86
  50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. Am Surg. 2008;74:1177-1181.
  51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. Clin Dermatol. 2017;35:225-227. doi:10.1016/j.clindermatol.2016.10.019
  52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. Clin Dermatol. 2014;32:592-598. doi:10.1016/j.clindermatol.2014.04.006
  53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. Dermatol Surg. 2019;45:519-526. doi:10.1097/DSS.0000000000001713
  54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. Medicine (Baltimore). 2017;96:e5857. doi:10.1097/MD.0000000000005857
  55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology. 2010;220:147-153. doi:10.1159/000269836
  56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. Dermatol Surg. 2017;43:107-115. doi:10.1097/DSS.0000000000000944
  57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. J Am Acad Dermatol. 2013;68:412-419. doi:10.1016/j.jaad.2012.07.027
  58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. Eur J Cancer. 2012;48:2046-2053. doi:10.1016/j.ejca.2012.01.003
  59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. Int Wound J. 2015;12:741-743. doi:10.1111/iwj.12206
  60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. J Cutan Pathol. 1992;19:116-123. doi:10.1111/j.1600-0560.1992.tb01352.x
  61. Montaudié H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis. 2016;11:117. doi:10.1186/s13023-016-0489-9.
  62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. Ann N Y Acad Sci. 2010;1194:213-222. doi:10.1111/j.1749-6632.2010.05463.x
  63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. Acta Derm Venereol. 2018;98:70-76. doi:10.2340/00015555-2781
  64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 1989;93:3-9. doi:10.1111/1523-1747.ep12277331
  65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522-3534. doi:10.1158/0008-5472.CAN-11-2996
  66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2004;123:788-790. doi:10.1111/j.0022-202X.2004.23418.x
  67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. Anal Cell Pathol (Amst). 2011;34:339-353. doi:10.3233/ACP-2011-0039
  68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. Br J Dermatol. 2008;158:778-785. doi:10.1111/j.1365-2133.2008.08466.x
  69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2007;16:465-467. doi:10.1111/j.1600-0625.2007.00563.x
  70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science. 2005;307:1773-1776. doi:10.1126/science.1106209
  71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol. 2014;170:1256-1265. doi:10.1111/bjd.12715
  72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2010;130:2853-2855. doi:10.1038/jid.2010.243
  73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. Dermatol Clin. 2010;28:171-178. doi:10.1016/j.det.2009.10.023
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.

Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.

Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).

Issue
Cutis - 113(1)
Publications
Topics
Page Number
29-34
Sections
Author and Disclosure Information

From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.

Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.

Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).

Author and Disclosure Information

From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.

Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.

Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).

Article PDF
Article PDF

As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.2 Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle.

Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.3 One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.

Discoid Lupus Erythematosus

Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,4 and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,5 India,6 and Japan7 have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,5 which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).5,8

Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.5 It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,9 as SCCs arising in DLE lesions virtually always display prominent solar elastosis6; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.10

Kuraitis_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Invasive%20squamous%20cell%20carcinoma%20arising%20within%20a%20labial%20discoid%20lupus%20erythematosus%20lesion.%20This%20patient%E2%80%99s%20lesions%20were%20present%20for%20approximately%206%20years%20prior%20to%20presentation%20for%20carcinoma.%3C%2Fp%3E

Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,11 thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.12 However, testing for HPV is not routinely performed in these cases.

Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips.

Lichen Planus

Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,13 variants of lichen planus may predispose patients to SCC.

 

 

Oral Lichen Planus—Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,14 compared to a reference rate of 0.2% for oral SCC.15 A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.16 The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.15 Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.17

Hypertrophic Lichen Planus—The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.13 A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.18

Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.19 In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.20

It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.21,22 In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment.

Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.23 Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.Nail Lichen Planus—Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.24 Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.25,26

Lichen Sclerosus

Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.27 It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N=78) may have adjacent LS.28

Kuraitis_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Poorly%20differentiated%20squamous%20cell%20carcinoma%20arising%20within%20vulvar%20lichen%20sclerosus.%20This%20patient%E2%80%99s%20dermatosis%20was%20present%20for%20approximately%207%20years%20prior%20to%20presentation%20for%20carcinoma.%3C%2Fp%3E

In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.29 Other studies have estimated a lag time of 4 years until SCC presentation.30 An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.31 It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.32 Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.33 Extragenital LS does not appear to have similar potential for malignant transformation.34

 

 

In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.35 Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.35 In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.36 These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.

The rate of SCC arising in male genital LS may approach 8.4%,37 with a lag time of 17 years from onset of LS to SCC diagnosis.38 Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.39 Male penile SCC in a background of LS may not necessarily be HPV associated.40

Marjolin Ulcer

Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.41 Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.42 Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.43 Cohort studies of MU on the lower extremities have observed lag times of 26.444 and 37.945 years, with both studies also noting relatively high rates of local recurrence.

The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,41 and local accumulation of toxin may promote mutagenesis.46 Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system47 and become even more aggressive as the tumor accumulates.48 Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.49

Hidradenitis Suppurativa

As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.50 Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.51 Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, β-defensins, and interleukins.52

A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.53 After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.53 A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.54 As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.55

Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),54,56 despite HS being more prevalent in females (2:1 ratio).57 Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women58; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.

 

 

Necrobiosis Lipoidica Diabeticorum

Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.59 The lag time was 21.5 years; 31% of cases (N=14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized.

Epidermolysis Bullosa

Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.60 Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.61 Although SCCs tend to be well differentiated in RDEB (73.9%),61 they also exhibit highly aggressive behavior.62 In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.63

As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.64 There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.65 Mutations in p53,66 local alterations in transforming growth factor β activity,67 and downstream matrix metalloproteinase activity68 have been implicated.

Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.69 Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.70 A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.71

There does not appear to be evidence for HPV involvement in RDEB-associated SCC.72 Squamous cell carcinoma development in RDEB appears to be multifactorial,73 but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.73

Conclusion

Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.


As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.2 Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle.

Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.3 One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.

Discoid Lupus Erythematosus

Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,4 and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,5 India,6 and Japan7 have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,5 which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).5,8

Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.5 It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,9 as SCCs arising in DLE lesions virtually always display prominent solar elastosis6; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.10

Kuraitis_1.jpg
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20Invasive%20squamous%20cell%20carcinoma%20arising%20within%20a%20labial%20discoid%20lupus%20erythematosus%20lesion.%20This%20patient%E2%80%99s%20lesions%20were%20present%20for%20approximately%206%20years%20prior%20to%20presentation%20for%20carcinoma.%3C%2Fp%3E

Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,11 thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.12 However, testing for HPV is not routinely performed in these cases.

Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips.

Lichen Planus

Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,13 variants of lichen planus may predispose patients to SCC.

 

 

Oral Lichen Planus—Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,14 compared to a reference rate of 0.2% for oral SCC.15 A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.16 The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.15 Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.17

Hypertrophic Lichen Planus—The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.13 A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.18

Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.19 In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.20

It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.21,22 In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment.

Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.23 Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.Nail Lichen Planus—Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.24 Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.25,26

Lichen Sclerosus

Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.27 It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N=78) may have adjacent LS.28

Kuraitis_2.jpg
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20Poorly%20differentiated%20squamous%20cell%20carcinoma%20arising%20within%20vulvar%20lichen%20sclerosus.%20This%20patient%E2%80%99s%20dermatosis%20was%20present%20for%20approximately%207%20years%20prior%20to%20presentation%20for%20carcinoma.%3C%2Fp%3E

In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.29 Other studies have estimated a lag time of 4 years until SCC presentation.30 An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.31 It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.32 Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.33 Extragenital LS does not appear to have similar potential for malignant transformation.34

 

 

In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.35 Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.35 In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.36 These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.

The rate of SCC arising in male genital LS may approach 8.4%,37 with a lag time of 17 years from onset of LS to SCC diagnosis.38 Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.39 Male penile SCC in a background of LS may not necessarily be HPV associated.40

Marjolin Ulcer

Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.41 Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.42 Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.43 Cohort studies of MU on the lower extremities have observed lag times of 26.444 and 37.945 years, with both studies also noting relatively high rates of local recurrence.

The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,41 and local accumulation of toxin may promote mutagenesis.46 Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system47 and become even more aggressive as the tumor accumulates.48 Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.49

Hidradenitis Suppurativa

As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.50 Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.51 Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, β-defensins, and interleukins.52

A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.53 After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.53 A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.54 As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.55

Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),54,56 despite HS being more prevalent in females (2:1 ratio).57 Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women58; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.

 

 

Necrobiosis Lipoidica Diabeticorum

Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.59 The lag time was 21.5 years; 31% of cases (N=14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized.

Epidermolysis Bullosa

Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.60 Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.61 Although SCCs tend to be well differentiated in RDEB (73.9%),61 they also exhibit highly aggressive behavior.62 In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.63

As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.64 There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.65 Mutations in p53,66 local alterations in transforming growth factor β activity,67 and downstream matrix metalloproteinase activity68 have been implicated.

Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.69 Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.70 A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.71

There does not appear to be evidence for HPV involvement in RDEB-associated SCC.72 Squamous cell carcinoma development in RDEB appears to be multifactorial,73 but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.73

Conclusion

Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.


References
  1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121:2373-2380. doi:10.1002/ijc.23173
  2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436-444. doi:10.1038/nature07205
  3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2011;2:98. doi:10.3389/fimmu.2011.00098
  4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. Clin Dermatol. 2004;22:121-124. doi:10.1016/j.clindermatol.2003.12.018
  5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. J Am Acad Dermatol. 2012;66:695-696. doi:10.1016 /j.jaad.2011.09.033
  6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. Lupus. 2015;24:1562-1566. doi:10.1177/0961203315599245
  7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. Eur J Dermatol. 2016;26:395-396. doi:10.1684/ejd.2016.2780
  8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. J Clin Oncol. 2005;23:759-765. doi:10.1200/JCO.2005.02.155
  9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol. 2018;125:e22-e30. doi:10.1016/j.oooo.2017.08.012
  10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. J Clin Rheumatol. 2011;17:35-36. doi:10.1097/RHU.0b013e3182051928
  11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol. 2015;27:454-460. doi:10.1097/BOR.0000000000000202
  12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. Cutis. 2010;86:195-198.
  13. Sigurgeirsson B, Lindelöf B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. Arch Dermatol. 1991;127:1684-1688. doi:10.1001/archderm.127.11.1684
  14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc. 2014;145:45-56. doi:10.14219/jada.2013.10
  15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol. 2019;58:296-301. doi:10.1111/ijd.14215
  16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol. 2017;68:92-102. doi:10.1016/j.oraloncology.2017.03.012
  17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. Odontology. 2017;105:262-266. doi:10.1007/s10266-016-0255-4
  18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. Dermatol Surg. 2015;41:1411-1418. doi:10.1097/DSS.0000000000000565
  19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. Pediatr Dermatol. 2015;32:e98-e101. doi:10.1111/pde.12549
  20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. Dermatol Online J. 2003;9:10.
  21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. J Cutan Pathol. 2012;39:243-250. doi:10.1111/j.1600-0560.2011.01834.x
  22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. Dermatol Surg. 2018;44:1466-1470. doi:10.1097/DSS.0000000000001465
  23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. JAAD Case Rep. 2017;3:151-154. doi: 10.1016/j.jdcr.2017.01.020
  24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. J Am Acad Dermatol. 2005;53:908-909. doi:10.1016/j.jaad.2005.04.052
  25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. J Am Acad Dermatol. 2011;64:1147-1153. doi:10.1016/j.jaad.2010.02.057
  26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. J Am Acad Dermatol. 2019;81:1358-1370. doi:10.1016/j.jaad.2019.03.070
  27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416. doi:10.1016/0190-9622(95)90060-8
  28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. Br J Obstet Gynaecol. 1990;97:1135-1139. doi:10.1111/j.1471-0528.1990.tb02502.x
  29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:1224-1230. doi:10.1158/1055-9965.EPI-16-0019
  30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol. 1998;29:932-948. doi:10.1016/s0046-8177(98)90198-8
  31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. J Low Genit Tract Dis. 2016;20:180-183. doi:10.1097/LGT.0000000000000186
  32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. JAMA Dermatol. 2015;151:1059-1060. doi:10.1001/jamadermatol.2015.0644
  33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. Am J Surg Pathol. 2018;42:828-835. doi:10.1097/PAS.0000000000001047
  34. Val I, Almeida G. An overview of lichen sclerosus. Clin Obstet Gynecol. 2005;48:808-817. doi:10.1097/01.grf.0000179635.64663.3d
  35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. JAMA Dermatol. 2015;151:1061-1067. doi:10.1001/jamadermatol.2015.0643
  36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. Arch Dermatol. 2004;140:709-712. doi:10.1001/archderm.140.6.709
  37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. Urology. 2010;76(2 suppl 1):S24-S35. doi:10.1016/j.urology.2010.04.007
  38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911-914. doi:10.1016/s0190-9622(99)70245-8
  39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. BJU Int. 2013;111:970-976. doi:10.1111/j.1464-410X.2012.11773.x
  40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. Am J Surg Pathol. 2003;27:1448-1453. doi:10.1097/00000478-200311000-00007
  41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64. doi:10.1016/j.jcws.2012.04.001
  42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? Burns. 2005;31:421-431. doi:10.1016/j.burns.2005.02.008
  43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? Int Wound J. 2019;16:479-485. doi:10.1111/iwj.13058
  44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. Ann Plast Surg. 2019;83:518-522. doi:10.1097/SAP.0000000000001995
  45. Karasoy Yesilada A, Zeynep Sevim K, Özgur Sucu D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. J Cutan Med Surg. 2013;17:404-409. doi:10.2310/7750.2013.13016
  46. Bazalin´ski D, Przybek-Mita J, Baran´ska B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. Contemp Oncol (Pozn). 2017;21:197-202. doi:10.5114/wo.2017.70109
  47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. Ann Plast Surg. 1986;16:42-48. doi:10.1097/00000637-198601000-00004
  48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? Plast Reconstr Surg. 1976;57:66-69.
  49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. Plast Reconstr Surg. 2009;123:184-191. doi:10.1097/PRS.0b013e3181904d86
  50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. Am Surg. 2008;74:1177-1181.
  51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. Clin Dermatol. 2017;35:225-227. doi:10.1016/j.clindermatol.2016.10.019
  52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. Clin Dermatol. 2014;32:592-598. doi:10.1016/j.clindermatol.2014.04.006
  53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. Dermatol Surg. 2019;45:519-526. doi:10.1097/DSS.0000000000001713
  54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. Medicine (Baltimore). 2017;96:e5857. doi:10.1097/MD.0000000000005857
  55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology. 2010;220:147-153. doi:10.1159/000269836
  56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. Dermatol Surg. 2017;43:107-115. doi:10.1097/DSS.0000000000000944
  57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. J Am Acad Dermatol. 2013;68:412-419. doi:10.1016/j.jaad.2012.07.027
  58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. Eur J Cancer. 2012;48:2046-2053. doi:10.1016/j.ejca.2012.01.003
  59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. Int Wound J. 2015;12:741-743. doi:10.1111/iwj.12206
  60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. J Cutan Pathol. 1992;19:116-123. doi:10.1111/j.1600-0560.1992.tb01352.x
  61. Montaudié H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis. 2016;11:117. doi:10.1186/s13023-016-0489-9.
  62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. Ann N Y Acad Sci. 2010;1194:213-222. doi:10.1111/j.1749-6632.2010.05463.x
  63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. Acta Derm Venereol. 2018;98:70-76. doi:10.2340/00015555-2781
  64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 1989;93:3-9. doi:10.1111/1523-1747.ep12277331
  65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522-3534. doi:10.1158/0008-5472.CAN-11-2996
  66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2004;123:788-790. doi:10.1111/j.0022-202X.2004.23418.x
  67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. Anal Cell Pathol (Amst). 2011;34:339-353. doi:10.3233/ACP-2011-0039
  68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. Br J Dermatol. 2008;158:778-785. doi:10.1111/j.1365-2133.2008.08466.x
  69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2007;16:465-467. doi:10.1111/j.1600-0625.2007.00563.x
  70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science. 2005;307:1773-1776. doi:10.1126/science.1106209
  71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol. 2014;170:1256-1265. doi:10.1111/bjd.12715
  72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2010;130:2853-2855. doi:10.1038/jid.2010.243
  73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. Dermatol Clin. 2010;28:171-178. doi:10.1016/j.det.2009.10.023
References
  1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. Int J Cancer. 2007;121:2373-2380. doi:10.1002/ijc.23173
  2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. Nature. 2008;454:436-444. doi:10.1038/nature07205
  3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. Front Immunol. 2011;2:98. doi:10.3389/fimmu.2011.00098
  4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. Clin Dermatol. 2004;22:121-124. doi:10.1016/j.clindermatol.2003.12.018
  5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. J Am Acad Dermatol. 2012;66:695-696. doi:10.1016 /j.jaad.2011.09.033
  6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. Lupus. 2015;24:1562-1566. doi:10.1177/0961203315599245
  7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. Eur J Dermatol. 2016;26:395-396. doi:10.1684/ejd.2016.2780
  8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. J Clin Oncol. 2005;23:759-765. doi:10.1200/JCO.2005.02.155
  9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol. 2018;125:e22-e30. doi:10.1016/j.oooo.2017.08.012
  10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. J Clin Rheumatol. 2011;17:35-36. doi:10.1097/RHU.0b013e3182051928
  11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. Curr Opin Rheumatol. 2015;27:454-460. doi:10.1097/BOR.0000000000000202
  12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. Cutis. 2010;86:195-198.
  13. Sigurgeirsson B, Lindelöf B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. Arch Dermatol. 1991;127:1684-1688. doi:10.1001/archderm.127.11.1684
  14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. J Am Dent Assoc. 2014;145:45-56. doi:10.14219/jada.2013.10
  15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. Int J Dermatol. 2019;58:296-301. doi:10.1111/ijd.14215
  16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. Oral Oncol. 2017;68:92-102. doi:10.1016/j.oraloncology.2017.03.012
  17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. Odontology. 2017;105:262-266. doi:10.1007/s10266-016-0255-4
  18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. Dermatol Surg. 2015;41:1411-1418. doi:10.1097/DSS.0000000000000565
  19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. Pediatr Dermatol. 2015;32:e98-e101. doi:10.1111/pde.12549
  20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. Dermatol Online J. 2003;9:10.
  21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. J Cutan Pathol. 2012;39:243-250. doi:10.1111/j.1600-0560.2011.01834.x
  22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. Dermatol Surg. 2018;44:1466-1470. doi:10.1097/DSS.0000000000001465
  23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. JAAD Case Rep. 2017;3:151-154. doi: 10.1016/j.jdcr.2017.01.020
  24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. J Am Acad Dermatol. 2005;53:908-909. doi:10.1016/j.jaad.2005.04.052
  25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. J Am Acad Dermatol. 2011;64:1147-1153. doi:10.1016/j.jaad.2010.02.057
  26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. J Am Acad Dermatol. 2019;81:1358-1370. doi:10.1016/j.jaad.2019.03.070
  27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am Acad Dermatol. 1995;32:393-416. doi:10.1016/0190-9622(95)90060-8
  28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. Br J Obstet Gynaecol. 1990;97:1135-1139. doi:10.1111/j.1471-0528.1990.tb02502.x
  29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2016;25:1224-1230. doi:10.1158/1055-9965.EPI-16-0019
  30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. Hum Pathol. 1998;29:932-948. doi:10.1016/s0046-8177(98)90198-8
  31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. J Low Genit Tract Dis. 2016;20:180-183. doi:10.1097/LGT.0000000000000186
  32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. JAMA Dermatol. 2015;151:1059-1060. doi:10.1001/jamadermatol.2015.0644
  33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. Am J Surg Pathol. 2018;42:828-835. doi:10.1097/PAS.0000000000001047
  34. Val I, Almeida G. An overview of lichen sclerosus. Clin Obstet Gynecol. 2005;48:808-817. doi:10.1097/01.grf.0000179635.64663.3d
  35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. JAMA Dermatol. 2015;151:1061-1067. doi:10.1001/jamadermatol.2015.0643
  36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. Arch Dermatol. 2004;140:709-712. doi:10.1001/archderm.140.6.709
  37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. Urology. 2010;76(2 suppl 1):S24-S35. doi:10.1016/j.urology.2010.04.007
  38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. J Am Acad Dermatol. 1999;41:911-914. doi:10.1016/s0190-9622(99)70245-8
  39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. BJU Int. 2013;111:970-976. doi:10.1111/j.1464-410X.2012.11773.x
  40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. Am J Surg Pathol. 2003;27:1448-1453. doi:10.1097/00000478-200311000-00007
  41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. J Am Col Certif Wound Spec. 2011;3:60-64. doi:10.1016/j.jcws.2012.04.001
  42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? Burns. 2005;31:421-431. doi:10.1016/j.burns.2005.02.008
  43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? Int Wound J. 2019;16:479-485. doi:10.1111/iwj.13058
  44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. Ann Plast Surg. 2019;83:518-522. doi:10.1097/SAP.0000000000001995
  45. Karasoy Yesilada A, Zeynep Sevim K, Özgur Sucu D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. J Cutan Med Surg. 2013;17:404-409. doi:10.2310/7750.2013.13016
  46. Bazalin´ski D, Przybek-Mita J, Baran´ska B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. Contemp Oncol (Pozn). 2017;21:197-202. doi:10.5114/wo.2017.70109
  47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. Ann Plast Surg. 1986;16:42-48. doi:10.1097/00000637-198601000-00004
  48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? Plast Reconstr Surg. 1976;57:66-69.
  49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. Plast Reconstr Surg. 2009;123:184-191. doi:10.1097/PRS.0b013e3181904d86
  50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. Am Surg. 2008;74:1177-1181.
  51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. Clin Dermatol. 2017;35:225-227. doi:10.1016/j.clindermatol.2016.10.019
  52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. Clin Dermatol. 2014;32:592-598. doi:10.1016/j.clindermatol.2014.04.006
  53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. Dermatol Surg. 2019;45:519-526. doi:10.1097/DSS.0000000000001713
  54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. Medicine (Baltimore). 2017;96:e5857. doi:10.1097/MD.0000000000005857
  55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology. 2010;220:147-153. doi:10.1159/000269836
  56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. Dermatol Surg. 2017;43:107-115. doi:10.1097/DSS.0000000000000944
  57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. J Am Acad Dermatol. 2013;68:412-419. doi:10.1016/j.jaad.2012.07.027
  58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. Eur J Cancer. 2012;48:2046-2053. doi:10.1016/j.ejca.2012.01.003
  59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. Int Wound J. 2015;12:741-743. doi:10.1111/iwj.12206
  60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. J Cutan Pathol. 1992;19:116-123. doi:10.1111/j.1600-0560.1992.tb01352.x
  61. Montaudié H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. Orphanet J Rare Dis. 2016;11:117. doi:10.1186/s13023-016-0489-9.
  62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. Ann N Y Acad Sci. 2010;1194:213-222. doi:10.1111/j.1749-6632.2010.05463.x
  63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. Acta Derm Venereol. 2018;98:70-76. doi:10.2340/00015555-2781
  64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. J Invest Dermatol. 1989;93:3-9. doi:10.1111/1523-1747.ep12277331
  65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. Cancer Res. 2012;72:3522-3534. doi:10.1158/0008-5472.CAN-11-2996
  66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2004;123:788-790. doi:10.1111/j.0022-202X.2004.23418.x
  67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. Anal Cell Pathol (Amst). 2011;34:339-353. doi:10.3233/ACP-2011-0039
  68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. Br J Dermatol. 2008;158:778-785. doi:10.1111/j.1365-2133.2008.08466.x
  69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. Exp Dermatol. 2007;16:465-467. doi:10.1111/j.1600-0625.2007.00563.x
  70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. Science. 2005;307:1773-1776. doi:10.1126/science.1106209
  71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. Br J Dermatol. 2014;170:1256-1265. doi:10.1111/bjd.12715
  72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. J Invest Dermatol. 2010;130:2853-2855. doi:10.1038/jid.2010.243
  73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. Dermatol Clin. 2010;28:171-178. doi:10.1016/j.det.2009.10.023
Issue
Cutis - 113(1)
Issue
Cutis - 113(1)
Page Number
29-34
Page Number
29-34
Publications
Publications
Topics
Article Type
Display Headline
Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses
Display Headline
Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>Kuraitis</fileName> <TBEID>0C02EE4B.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EE4B</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Kuraitis</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240108T074505</firstPublished> <LastPublished>20240108T074505</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240108T074505</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Drew Kuraitis, MD, PhD; Andrea Murina, MD</byline> <bylineText>Drew Kuraitis, MD, PhD; Andrea Murina, MD</bylineText> <bylineFull>Drew Kuraitis, MD, PhD; Andrea Murina, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>29-34</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.1 Extrinsic inflammation leads to generation of proinfla</metaDescription> <articlePDF>299909</articlePDF> <teaserImage/> <title>Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>nonmelanoma skin cancer</keyword> <keyword> dermatopathology</keyword> <keyword> squamous cell carcinoma</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">245</term> <term>204</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026a0.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Squamous Cell Carcinoma Arising in Chronic Inflammatory Dermatoses</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Squamous cell carcinoma (SCC) is a known sequela of chronic inflammatory conditions of the skin. Labial discoid lupus erythematosus (DLE), oral lichen planus (OLP), and lichen sclerosus have a relatively short lag time from dermatosis onset to manifestation of malignancy; cutaneous DLE, hypertrophic lichen planus, chronic wounds, hidradenitis suppurativa (HS), and necrobiosis lipoidica can be present for decades before an associated malignancy is observed. Vigilant monitoring is essential for orolabial DLE, chronic HS, and chronic wounds because malignancies in these settings are particularly aggressive and often fatal. We summarize what is known about the nature and demographics of SCC arising within chronic inflammatory dermatoses, emphasizing lag time from dermatosis diagnosis to malignancy onset of common inflammatory conditions.</p> <p> <em><em>Cutis.</em> 2024;113:29-34.</em> </p> <p>As many as one-quarter of human cancers are related to chronic inflammation, chronic infection, or both.<sup>1</sup> Extrinsic inflammation leads to generation of proinflammatory cytokines that in turn recruit other inflammatory cells, which is thought to generate a positive amplification loop.<sup>2</sup> Intrinsic stimuli from proto-oncogenes and mutations in tumor suppressor genes lead to transformed cancer cells that also secrete proinflammatory cytokines, thus propagating the cycle. </p> <p>Numerous factors have been observed in association with tumor growth, progression, invasion, and metastasis.<sup>3</sup> One factor for the development of squamous cell carcinoma (SCC) may be chronic inflammatory dermatoses. To date, reviews of chronic inflammation–associated malignancy have focused on solid organ cancers. We sought to provide an up-to-date review of SCC arising within chronic dermatoses, with an emphasis on the anatomic location of dermatoses involved in the transformation of cancer cells, the lag time from onset of dermatosis to diagnosis of SCC, and the distinctive mechanisms thought to be involved in the tumorigenesis in particular dermatoses.</p> <h3>Discoid Lupus Erythematosus</h3> <p>Discoid lupus erythematosus (DLE) is a chronic cutaneous lupus erythematosus variant with a female to male predominance of 3:1,<sup>4</sup> and DLE lesions are prone to malignant transformation. Retrospective cohort studies have attempted to characterize who is at risk for SCC and how SCCs behave depending on their location. Cohorts from China,<sup>5</sup> India,<sup>6</sup> and Japan<sup>7</sup> have noted a higher rate of SCC within DLE lesions in men (female to male ratios of 1:2.2, 1:1.6, and 1:2, respectively) and shorter lag times for SCC onset within DLE lesions of the lips (13, 5, and 10 years, respectively) compared to SCC arising in DLE elsewhere (19.2, 11.2, and 26 years, respectively). Studies have noted that DLE lesions of the lips may be prone to more rapid SCC tumorigenesis compared to DLE on cutaneous sites. One study reported SCC in DLE recurrence, metastasis, and death rates of 29%, 16.1%, and 19.4%, respectively,<sup>5</sup> which exceeds reported rates in non-DLE SCCs (20%, 0.5% to 6%, and 1%, respectively).<sup>5,8</sup></p> <p>Because SCC arising within DLE is most common on the lips (Figure 1), it has been hypothesized that the high rate of transformation of DLE lesions on the lips may be due to constant exposure to irritation and tobacco, which may accelerate carcinogenesis.<sup>5</sup> It also has been hypothesized that atrophic discoid lesions have lost sun protection and are more prone to mutagenic UV radiation,<sup>9</sup> as SCCs arising in DLE lesions virtually always display prominent solar elastosis<sup>6</sup>; however, SCC has been observed to arise in non–sun-exposed DLE lesions in both White and Black patients.<sup>10<br/><br/></sup>Additionally, use of immunosuppressant medications may accelerate the emergence of malignancy or more aggressive forms of malignancy; however, patients with autoimmune disease have a greater risk for malignancy at baseline,<sup>11</sup> thus making it difficult to determine the excess risk from medications. There also may be a role for human papillomavirus (HPV) accelerating SCC development in DLE lesions, as demonstrated in a case of SCC arising in DLE lesions of the ears, with viral staining evident within the tumors.<sup>12</sup> However, testing for HPV is not routinely performed in these cases. <br/><br/>Dermatologists need to be aware of the relatively rapid tumorigenesis and aggressive behavior of transformation and aggression seen with SCC arising within orolabial DLE lesions compared to cutaneous lesions, especially those on the lips. </p> <h3>Lichen Planus</h3> <p>Although patients with typical cutaneous lichen planus lesions do not have an increased risk for SCC,<sup>13</sup> variants of lichen planus may predispose patients to SCC.</p> <p><i>Oral Lichen Planus—</i>Oral lichen planus (OLP) lesions are prone to malignant transformation. A systematic review of 16 studies evaluating the risk for OLP-associated SCC revealed an overall transformation rate of 1.09%, with a mean lag time of 4.3 years,<sup>14</sup> compared to a reference rate of 0.2% for oral SCC.<sup>15</sup> A meta-analysis of 19,676 patients with OLP and other oral lichenoid lesions revealed an oral SCC rate of 1.1%, with higher rates of transformation seen in cigarette smokers, alcoholics, and patients with hepatitis C virus infection.<sup>16</sup> The ulcerative subtype of OLP appears to present a greater risk for malignant transformation.<sup>15</sup> Dermatologists also should be cognizant that treatments for OLP such as topical calcineurin inhibitors may support the development of malignancy within inflammatory lesions.<sup>17<br/><br/></sup><i>Hypertrophic Lichen Planus—</i>The hypertrophic variant of lichen planus (HLP) also is prone to malignant transformation. A 1991 epidemiologic study from Sweden of malignancy arising in lichen planus revealed a disproportionate number of cases arising in verrucous or hypertrophic lesions, with a mean of 12.2 years from onset of the dermatosis to malignancy diagnosis.<sup>13</sup> A subsequent 2015 retrospective study of 38 patients revealed that SCC had a propensity for the lower limb, favoring the pretibial region and the calf over the foot and the ankle with a reported lag time of 11 years.<sup>18<br/><br/></sup>Although metastatic SCC arising in HLP is rare, 2 cases have been reported. A 24-year-old woman presented with an HLP plaque on the lower leg that developed during childhood and rapidly enlarged 2 months prior to presentation; she eventually died from metastatic disease.<sup>19</sup> In another case, a 34-year-old man presented with an HLP lesion of approximately 10 years’ duration. A well-differentiated SCC was excised, and he developed lymph node metastases 5 months later.<sup>20<br/><br/></sup>It is important to note that HLP on the legs often is misdiagnosed as SCC, as pseudoepitheliomatous hyperplasia and squamous metaplasia can be difficult to differentiate clinically and histologically.<sup>21,22</sup> In the case of multiple eruptive SCCs of the lower leg, clinical correlation is essential to avoid unnecessary and ineffective surgical treatment. <br/><br/>Patients with HLP may exhibit Wickham striae, follicular accentuation, and mucocutaneous lichen planus at other sites, or a correlative initiation of possible culprit medications.<sup>23</sup> Because true SCC arising within HLP is relatively rare, its malignant potential is not as clear as those arising within DLE; however, the lower limb appears to be the most common location for SCC within HLP.<i>Nail Lichen Planus—</i>Squamous cell carcinoma arising in nail lichen planus is rare. A report of 2 patients were diagnosed with lichen planus approximately 15 years prior to diagnosis of ungual SCC.<sup>24</sup> Given the rarity of this presentation, it is difficult to ascertain the approximate lag time and other risk factors. Furthermore, the role of HPV in these cases was not ruled out. Oncogenic HPV strains have been reported in patients with periungual SCC.<sup>25,26</sup></p> <h3>Lichen Sclerosus</h3> <p>Lichen sclerosus (LS) is a chronic inflammatory dermatosis that favors the anogenital area in a female to male ratio of 10:1.<sup>27</sup> It is considered a premalignant condition for SCC tumorigenesis and may be a strong predictor of vulvar SCC (Figure 2), as 62% of vulvar SCC cases (N<span class="body">=</span>78) may have adjacent LS.<sup>28</sup></p> <p>In a Dutch cohort of 3038 women with LS, 2.6% of patients developed vulvar SCC at a median of 3.3 years after LS diagnosis.<sup>29</sup> Other studies have estimated a lag time of 4 years until SCC presentation.<sup>30</sup> An Italian cohort of 976 women similarly observed that 2.7% of patients developed premalignancy or SCC.<sup>31</sup> It was previously estimated that 3% to 5% of patients with LS developed SCC; however, prior studies may have included cases of vulvar intraepithelial neoplasia with low risk for invasive SCC, which might have overestimated true risk of SCC.<sup>32</sup> Another confounding factor for elucidating SCC on a background of LS may be the presence of HPV.<sup>33</sup> Extragenital LS does not appear to have similar potential for malignant transformation.<sup>34<br/><br/></sup>In a prospective Australian cohort of 507 women with LS (mean age, 55.4 years), remission was induced with potent topical corticosteroids.<sup>35</sup> Patients who were adherent to a topical regimen did not develop SCC during follow-up. Those who were nonadherent or partially adherent had a 4.7% risk for SCC.<sup>35</sup> In a similar prospective study of 83 women in France, the SCC rate was 9.6% in lesions that were untreated or irregularly treated.<sup>36</sup> These studies provide essential evidence that appropriately treating LS can prevent SCC at a later date, though longer-term data are lacking.<br/><br/>The rate of SCC arising in male genital LS may approach 8.4%,<sup>37</sup> with a lag time of 17 years from onset of LS to SCC diagnosis.<sup>38</sup> Although circumcision often is considered curative for male genital LS, patients have been observed to develop penile SCC at least 5 years after circumcision.<sup>39</sup> Male penile SCC in a background of LS may not necessarily be HPV associated.<sup>40</sup></p> <h3>Marjolin Ulcer</h3> <p>Chronic ulcers or scars, typically postburn scars, may undergo malignant transformation, with SCC being the most common carcinoma.<sup>41</sup> Squamous cell carcinoma in the context of a chronic ulcer or wound is known as a Marjolin ulcer (MU). Up to 2% of burn scars have been observed to undergo malignant transformation.<sup>42</sup> Marjolin ulcers tend to behave aggressively once they form, and it has been proposed that removal of scar tissue may be a preventive therapeutic strategy.<sup>43</sup> Cohort studies of MU on the lower extremities have observed lag times of 26.4<sup>44 </sup>and 37.9<sup>45</sup> years, with both studies also noting relatively high rates of local recurrence.</p> <p>The pathogenesis of MU appears to be multifactorial. Chronic inflammation and scar formation have been implicated. Chronic inflammation and irritation of lesions at natural creases are thought to increase mitotic activity,<sup>41</sup> and local accumulation of toxin may promote mutagenesis.<sup>46</sup> Scar formation may create a locally immunoprivileged site, allowing for developing tumors to evade the immune system<sup>47</sup> and become even more aggressive as the tumor accumulates.<sup>48</sup> Scar formation also may prevent the ability of immune cells to penetrate the tumor microenvironment and access lymphatic channels.<sup>49</sup> </p> <h3>Hidradenitis Suppurativa</h3> <p>As many as 3.2% of patients with chronic hidradenitis suppurativa (HS) experience malignant transformation to SCC.<sup>50</sup> Early HS displays subclinical lymphedema in affected sites, which can progress to chronic fibrosis, stasis, and accumulation of protein-rich fluid.<sup>51</sup> Stasis changes have been associated with altered local inflammatory proteins, such as toll-like receptors, <span class="body">β</span>-defensins, and interleukins.<sup>52</sup> </p> <p>A retrospective cohort study of 12 patients revealed a lag time of 28.5 years from HS diagnosis to the manifestation of malignancy.<sup>53</sup> After local excision, 7 patients developed recurrence, with 100% mortality. Squamous cell carcinomas were well differentiated and moderately differentiated.<sup>53</sup> A 2017 literature review of 62 case reports calculated a mean lag time of 27 years. Despite 85% of SCCs being well differentiated and moderately differentiated, nearly half of patients died within 2 years.<sup>54</sup> As seen in other inflammatory conditions, HPV can complicate perineal HS and promote SCC tumorigenesis.<sup>55<br/><br/></sup>Squamous cell carcinomas arising within HS lesions are more prevalent in males (6.75:1 ratio),<sup>54,56</sup> despite HS being more prevalent in females (2:1 ratio).<sup>57</sup> Similar to DLE, SCCs arising in HS are aggressive and are seen more in males, despite both conditions being female predominant. Incidence and mortality rates for primary cutaneous SCC are higher for men vs women<sup>58</sup>; however, the discordance in aggressive behavior seen more commonly in SCC arising from HS or DLE in male patients has yet to be explained.</p> <h3>Necrobiosis Lipoidica Diabeticorum</h3> <p>Malignancy arising within necrobiosis lipoidica diabeticorum (NLD) is rare. A review of 14 published cases noted that 13 were SCC and 1 was leiomyosarcoma.<sup>59</sup> The lag time was 21.5 years; 31% of cases (N<span class="body">=</span>14) presented with regional lymph node metastasis. Although chronic ulceration is a risk factor for SCC and occurs in as many as one-third of NLD cases, its correlation with ulceration and malignant transformation has not been characterized. </p> <h3>Epidermolysis Bullosa</h3> <p>Recessive dystrophic epidermolysis bullosa (RDEB) is a noninflammatory inherited blistering disease, and patients have an inherently high risk for aggressive SCC.<sup>60</sup> Other forms of epidermolysis bullosa can lead to SCC, but the rarer RDEB accounts for 69% of SCC cases, with a median age of 36 years at presentation.<sup>61</sup> Although SCCs tend to be well differentiated in RDEB (73.9%),<sup>61</sup> they also exhibit highly aggressive behavior.<sup>62</sup> In the most severe variant—RDEB-generalized severe—the cumulative risk for SCC-related death in an Australian population was 84.4% at 34 years of age.<sup>63</sup> </p> <p>As RDEB is an inherited disorder with potential for malignancy at a young age, the pathogenesis is plausibly different from the previously discussed inflammatory dermatoses. This disease is characterized by a mutation in the collagen VII gene, leading to loss of anchoring fibrils and a basement membrane zone split.<sup>64</sup> There also can be inherent fibroblast alterations; RDEB fibroblasts create an environment for tumor growth by supporting malignant-cell adhesion and invasion.<sup>65</sup> Mutations in p53,<sup>66</sup> local alterations in transforming growth factor β activity,<sup>67</sup> and downstream matrix metalloproteinase activity<sup>68</sup> have been implicated. <br/><br/>Additionally, keratinocytes may retain the N-terminal noncollagenous (NC1) domain of truncated collagen VII while losing the anchoring NC2 domain in mutated collagen VII RDEB, thereby supporting anchorless keratinocyte survival and higher metastatic potential.<sup>69</sup> Retention of this truncated NC1 domain has shown conversion of RDEB keratinocytes to tumor in a xenotransplant mouse model.<sup>70</sup> A high level of type VII collagen itself may inherently be protumorigenic for keratinocytes.<sup>71</sup> <br/><br/>There does not appear to be evidence for HPV involvement in RDEB-associated SCC.<sup>72</sup> Squamous cell carcinoma development in RDEB appears to be multifactorial,<sup>73</sup> but validated tumor models are lacking. Other than conventional oncologic therapy, future directions in the management of RDEB may include gene-, protein- and cell-targeted therapies.<sup>73</sup></p> <h3>Conclusion</h3> <p>Squamous cell carcinomas are known to arise within chronic cutaneous inflammatory dermatoses. Tumorigenesis peaks relatively early in new orolabial DLE, LS, and OLP cases, and can occur over many decades in cutaneous DLE, HLP, HS, NLD, and chronic wounds or scars, summarized in the Table. Frequent SCCs are observed in high-risk subtypes of epidermolysis bullosa. Dermatologists must examine areas affected by these diseases at regular intervals, being mindful of the possibility of SCC development. Furthermore, dermatologists should adopt a lower threshold to biopsy suspicious lesions, especially those that develop within relatively new orolabial DLE, chronic HS, or chronic wound cases, as SCC in these settings is particularly aggressive and displays mortality and metastasis rates that exceed those of common cutaneous SCC.
</p> <h2>REFERENCES</h2> <p class="reference"> 1. Hussain SP, Harris CC. Inflammation and cancer: an ancient link with novel potentials. <i>Int J Cancer. </i>2007;121:2373-2380. <span class="citation-doi">doi:10.1002/ijc.23173<br/><br/></span> 2. Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation. <i>Nature. </i>2008;454:436-444. <span class="citation-doi">doi:10.1038/nature07205<br/><br/></span> 3. Multhoff G, Molls M, Radons J. Chronic inflammation in cancer development. <i>Front Immunol. </i>2011;2:98. <span class="citation-doi">doi:10.3389/fimmu.2011.00098<br/><br/></span> 4. Tebbe B. Clinical course and prognosis of cutaneous lupus erythematosus. <i>Clin Dermatol. </i>2004;22:121-124. <span class="citation-doi">doi:10.1016/j.clindermatol.2003.12.018</span> 5. Tao J, Zhang X, Guo N, et al. Squamous cell carcinoma complicating discoid lupus erythematosus in Chinese patients: review of the literature, 1964-2010. <i>J Am Acad Dermatol. </i>2012;66:695-696. <span class="citation-doi">doi:10.1016 /j.jaad.2011.09.033<br/><br/></span> 6. Fernandes MS, Girisha BS, Viswanathan N, et al. Discoid lupus erythematosus with squamous cell carcinoma: a case report and review of the literature in Indian patients. <i>Lupus. </i>2015;24:1562-1566. <span class="citation-doi">doi:10.1177/0961203315599245<br/><br/></span> 7. Makita E, Akasaka E, Sakuraba Y, et al. Squamous cell carcinoma on the lip arising from discoid lupus erythematosus: a case report and review of Japanese patients. <i>Eur J Dermatol. </i>2016;26:395-396. <span class="citation-doi">doi:10.1684/ejd.2016.2780<br/><br/></span> 8. Clayman GL, Lee JJ, Holsinger FC, et al. Mortality risk from squamous cell skin cancer. <i>J Clin Oncol. </i>2005;23:759-765. <span class="citation-doi">doi:10.1200/JCO.2005.02.155<br/><br/></span> 9. Arvanitidou I-E, Nikitakis NG, Georgaki M, et al. Multiple primary squamous cell carcinomas of the lower lip and tongue arising in discoid lupus erythematosus: a case report. <i>Oral Surg Oral Med Oral Pathol Oral Radiol. </i>2018;125:e22-e30. <span class="citation-doi">doi:10.1016/j.oooo.2017.08.012<br/><br/></span>10. Alsanafi S, Werth VP. Squamous cell carcinomas arising in discoid lupus erythematosus scars: unusual occurrence in an African-American and in a sun-protected area. <i>J Clin Rheumatol. </i>2011;17:35-36. <span class="citation-doi">doi:10.1097/RHU.0b013e3182051928<br/><br/></span>11. Goobie GC, Bernatsky S, Ramsey-Goldman R, et al. Malignancies in systemic lupus erythematosus: a 2015 update. <i>Curr Opin Rheumatol. </i>2015;27:454-460. <span class="citation-doi">doi:10.1097/BOR.0000000000000202<br/><br/></span>12. Simpson JK, Medina-Flores R, Deng J-S. Squamous cell carcinoma arising in discoid lupus erythematosus lesions of the ears infected with human papillomavirus. <i>Cutis. </i>2010;86:195-198.<br/><br/>13. Sigurgeirsson B, <span class="authors-list-item">Lindelöf </span>B. Lichen planus and malignancy. an epidemiologic study of 2071 patients and a review of the literature. <i>Arch Dermatol. </i>1991;127:1684-1688. <span class="citation-doi">doi:10.1001/archderm.127.11.1684<br/><br/></span>14. Fitzpatrick SG, Hirsch SA, Gordon SC. The malignant transformation of oral lichen planus and oral lichenoid lesions: a systematic review. <i>J Am Dent Assoc. </i>2014;145:45-56. <span class="citation-doi">doi:10.14219/jada.2013.10<br/><br/></span>15. Laniosz V, Torgerson RR, Ramos-Rodriguez AJ, et al. Incidence of squamous cell carcinoma in oral lichen planus: a 25-year population-based study. <i>Int J Dermatol. </i>2019;58:296-301. <span class="citation-doi">doi:10.1111/ijd.14215<br/><br/></span>16. Aghbari SMH, Abushouk AI, Attia A, et al. Malignant transformation of oral lichen planus and oral lichenoid lesions: a meta-analysis of 20095 patient data. <i>Oral Oncol. </i>2017;68:92-102. <span class="citation-doi">doi:10.1016/j.oraloncology.2017.03.012<br/><br/></span>17. Morita M, Asoda S, Tsunoda K, et al. The onset risk of carcinoma in patients continuing tacrolimus topical treatment for oral lichen planus: a case report. <i>Odontology. </i>2017;105:262-266. <span class="citation-doi">doi:10.1007/s10266-016-0255-4<br/><br/></span>18. Knackstedt TJ, Collins LK, Li Z, et al. Squamous cell carcinoma arising in hypertrophic lichen planus: a review and analysis of 38 cases. <i>Dermatol Surg. </i>2015;41:1411-1418. <span class="citation-doi">doi:10.1097/DSS.0000000000000565<br/><br/></span>19. Tong LX, Weinstock MJ, Drews R, et al. Widely metastatic squamous cell carcinoma originating from malignant transformation of hypertrophic lichen planus in a 24-year-old woman: case report and review of the literature. <i>Pediatr Dermatol. </i>2015;32:e98-e101. <span class="citation-doi">doi:10.1111/pde.12549<br/><br/></span>20. Ardabili M, Gambichler T, Rotterdam S, et al. Metastatic cutaneous squamous cell carcinoma arising from a previous area of chronic hypertrophic lichen planus. <i>Dermatol Online J. </i>2003;9:10.<br/><br/>21. Bowen AR, Burt L, Boucher K, et al. Use of proliferation rate, p53 staining and perforating elastic fibers in distinguishing keratoacanthoma from hypertrophic lichen planus: a pilot study. <i>J Cutan Pathol. </i>2012;39:243-250. <span class="citation-doi">doi:10.1111/j.1600-0560.2011.01834.x<br/><br/></span>22. Totonchy MB, Leventhal JS, Ko CJ, et al. Hypertrophic lichen planus and well-differentiated squamous cell carcinoma: a diagnostic conundrum. <i>Dermatol Surg. </i>2018;44:1466-1470. <span class="citation-doi">doi:10.1097/DSS.0000000000001465</span></p> <p class="reference">23. Levandoski KA, Nazarian RM, Asgari MM. Hypertrophic lichen planus mimicking squamous cell carcinoma: the importance of clinicopathologic correlation. <i>JAAD Case Rep. </i>2017;3:151-154. <span class="citation-doi">doi: 10.1016/j.jdcr.2017.01.020<br/><br/></span>24. Okiyama N, Satoh T, Yokozeki H, et al. Squamous cell carcinoma arising from lichen planus of nail matrix and nail bed. <i>J Am Acad Dermatol. </i>2005;53:908-909. <span class="citation-doi">doi:10.1016/j.jaad.2005.04.052<br/><br/></span>25. Riddel C, Rashid R, Thomas V. Ungual and periungual human papillomavirus-associated squamous cell carcinoma: a review. <i>J Am Acad Dermatol. </i>2011;64:1147-1153. doi:<span class="citation-doi">10.1016/j.jaad.2010.02.057<br/><br/></span>26. Shimizu A, Kuriyama Y, Hasegawa M, et al. Nail squamous cell carcinoma: a hidden high-risk human papillomavirus reservoir for sexually transmitted infections. <i>J Am Acad Dermatol. </i>2019;81:1358-1370. <span class="citation-doi">doi:10.1016/j.jaad.2019.03.070<br/><br/></span>27. Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. <i>J Am Acad Dermatol. </i>1995;32:393-416. <span class="citation-doi">doi:10.1016/0190-9622(95)90060-8<br/><br/></span>28. Leibowitch M, Neill S, Pelisse M, et al. The epithelial changes associated with squamous cell carcinoma of the vulva: a review of the clinical, histological and viral findings in 78 women. <i>Br J Obstet Gynaecol. </i>1990;97:1135-1139. <span class="citation-doi">doi:10.1111/j.1471-0528.1990.tb02502.x<br/><br/></span>29. Bleeker MCG, Visser PJ, Overbeek LIH, et al. Lichen sclerosus: incidence and risk of vulvar squamous cell carcinoma. <i>Cancer Epidemiol Biomarkers Prev. </i>2016;25:1224-1230. <span class="citation-doi">doi:10.1158/1055-9965.EPI-16-0019<br/><br/></span>30. Carlson JA, Ambros R, Malfetano J, et al. Vulvar lichen sclerosus and squamous cell carcinoma: a cohort, case control, and investigational study with historical perspective; implications for chronic inflammation and sclerosis in the development of neoplasia. <i>Hum Pathol. </i>1998;29:932-948. <span class="citation-doi">doi:10.1016/s0046-8177(98)90198-8<br/><br/></span>31. Micheletti L, Preti M, Radici G, et al. Vulvar lichen sclerosus and neoplastic transformation: a retrospective study of 976 cases. <i>J Low Genit Tract Dis. </i>2016;20:180-183. <span class="citation-doi">doi:10.1097/LGT.0000000000000186<br/><br/></span>32. Cooper SM, Madnani N, Margesson L. Reduced risk of squamous cell carcinoma with adequate treatment of vulvar lichen sclerosus. <i>JAMA Dermatol. </i>2015;151:1059-1060. <span class="citation-doi">doi:10.1001/jamadermatol.2015.0644<br/><br/></span>33. Rakislova N, Alemany L, Clavero O, et al; VVAP Study Group. Differentiated vulvar intraepithelial neoplasia-like and lichen sclerosus-like lesions in HPV-associated squamous cell carcinomas of the vulva. <i>Am J Surg Pathol. </i>2018;42:828-835. <span class="citation-doi">doi:10.1097/PAS.0000000000001047<br/><br/></span>34. Val I, Almeida G. An overview of lichen sclerosus. <i>Clin Obstet Gynecol. </i>2005;48:808-817. <span class="citation-doi">doi:10.1097/01.grf.0000179635.64663.3d<br/><br/></span>35. Lee A, Bradford J, Fischer G. Long-term management of adult vulvar lichen sclerosus: a prospective cohort study of 507 women. <i>JAMA Dermatol. </i>2015;151:1061-1067. <span class="citation-doi">doi:10.1001/jamadermatol.2015.0643</span></p> <p class="reference">36. Renaud-Vilmer C, Cavelier-Balloy B, Porcher R, et al. Vulvar lichen sclerosus: effect of long-term topical application of a potent steroid on the course of the disease. <i>Arch Dermatol. </i>2004;140:709-712. <span class="citation-doi">doi:10.1001/archderm.140.6.709<br/><br/></span>37. Minhas S, Manseck A, Watya S, et al. Penile cancer—prevention and premalignant conditions. <i>Urology. </i>2010;76(2 suppl 1):S24-S35. <span class="citation-doi">doi:10.1016/j.urology.2010.04.007<br/><br/></span>38. Nasca MR, Innocenzi D, Micali G. Penile cancer among patients with genital lichen sclerosus. <i>J Am Acad Dermatol. </i>1999;41:911-914. <span class="citation-doi">doi:10.1016/s0190-9622(99)70245-8<br/><br/></span>39. Philippou P, Shabbir M, Ralph DJ, et al. Genital lichen sclerosus/balanitis xerotica obliterans in men with penile carcinoma: a critical analysis. <i>BJU Int. </i>2013;111:970-976. <span class="citation-doi">doi:10.1111/j.1464-410X.2012.11773.x<br/><br/></span>40. Velazquez EF, Cubilla AL. Lichen sclerosus in 68 patients with squamous cell carcinoma of the penis: frequent atypias and correlation with special carcinoma variants suggests a precancerous role. <i>Am J Surg Pathol. </i>2003;27:1448-1453. <span class="citation-doi">doi:10.1097/00000478-200311000-00007<br/><br/></span>41. Pekarek B, Buck S, Osher L. A comprehensive review on Marjolin’s ulcers: diagnosis and treatment. <i>J Am Col Certif Wound Spec. </i>2011;3:60-64. <span class="citation-doi">doi:10.1016/j.jcws.2012.04.001<br/><br/></span>42. Aydogdu E, Yildirim S, Akoz T. Is surgery an effective and adequate treatment in advanced Marjolin’s ulcer? <i>Burns. </i>2005;31:421-431. <span class="citation-doi">doi:10.1016/j.burns.2005.02.008<br/><br/></span>43. Xiao H, Deng K, Liu R, et al. A review of 31 cases of Marjolin’s ulcer on scalp: is it necessary to preventively remove the scar? <i>Int Wound J. </i>2019;16:479-485. <span class="citation-doi">doi:10.1111/iwj.13058<br/><br/></span>44. Chaturvedi G, Gupta AK, Das S, et al. Marjolin ulcer: an observational epidemiological study from a tertiary care centre in India. <i>Ann Plast Surg. </i>2019;83:518-522. <span class="citation-doi">doi:10.1097/SAP.0000000000001995<br/><br/></span>45. Karasoy Yesilada A, Zeynep Sevim K, <span class="authors-list-item">Özgur Sucu </span>D, et al. Marjolin ulcer: clinical experience with 34 patients over 15 years. <i>J Cutan Med Surg. </i>2013;17:404-409. <span class="citation-doi">doi:10.2310/7750.2013.13016<br/><br/></span>46. <span class="authors-list-item">Bazali</span><span class="authors-list-item">n</span><span class="authors-list-item">´</span><span class="authors-list-item">ski</span> D, Przybek-Mita J, <span class="authors-list-item">Bara</span><span class="authors-list-item">n</span><span class="authors-list-item">´</span><span class="authors-list-item">ska</span> B, et al. Marjolin’s ulcer in chronic wounds - review of available literature. <i>Contemp Oncol (Pozn). </i>2017;21:197-202. <span class="citation-doi">doi:10.5114/wo.2017.70109<br/><br/></span>47. Visuthikosol V, Boonpucknavig V, Nitiyanant P. Squamous carcinoma in scars: clinicopathological correlations. <i>Ann Plast Surg. </i>1986;16:42-48. <span class="citation-doi">doi:10.1097/00000637-198601000-00004<br/><br/></span>48. Bostwick J 3rd, Pendergrast WJ Jr, Vasconez LO. Marjolin’s ulcer: an immunologically privileged tumor? <i>Plast Reconstr Surg. </i>1976;57:66-69.<br/><br/>49. Kerr-Valentic MA, Samimi K, Rohlen BH, et al. Marjolin’s ulcer: modern analysis of an ancient problem. <i>Plast Reconstr Surg. </i>2009;123:184-191. <span class="citation-doi">doi:10.1097/PRS.0b013e3181904d86<br/><br/></span>50. Constantinou C, Widom K, Desantis J, et al. Hidradenitis suppurativa complicated by squamous cell carcinoma. <i>Am Surg. </i>2008;74:1177-1181.<br/><br/>51. Fabbrocini G, Ruocco E, De Vita V, et al. Squamous cell carcinoma arising in long-standing hidradenitis suppurativa: an overlooked facet of the immunocompromised district. <i>Clin Dermatol. </i>2017;35:225-227. <span class="citation-doi">doi:10.1016/j.clindermatol.2016.10.019<br/><br/></span>52. Baroni A, Buommino E, Piccolo V, et al. Alterations of skin innate immunity in lymphedematous limbs: correlations with opportunistic diseases. <i>Clin Dermatol. </i>2014;32:592-598. <span class="citation-doi">doi:10.1016/j.clindermatol.2014.04.006<br/><br/></span>53. Kohorst JJ, Shah KK, Hallemeier CL, et al. Squamous cell carcinoma in perineal, perianal, and gluteal hidradenitis suppurativa: experience in 12 patients. <i>Dermatol Surg. </i>2019;45:519-526. <span class="citation-doi">doi:10.1097/DSS.0000000000001713<br/><br/></span>54. Huang C, Lai Z, He M, et al. Successful surgical treatment for squamous cell carcinoma arising from hidradenitis suppurativa: a case report and literature review. <i>Medicine (Baltimore). </i>2017;96:e5857. <span class="citation-doi">doi:10.1097/MD.0000000000005857<br/><br/></span>55. Lavogiez C, Delaporte E, Darras-Vercambre S, et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. <i>Dermatology. </i>2010;220:147-153. <span class="citation-doi">doi:10.1159/000269836<br/><br/></span>56. Makris G-M, Poulakaki N, Papanota A-M, et al. Vulvar, perianal and perineal cancer after hidradenitis suppurativa: a systematic review and pooled analysis. <i>Dermatol Surg. </i>2017;43:107-115. <span class="citation-doi">doi:10.1097/DSS.0000000000000944<br/><br/></span>57. Cosmatos I, Matcho A, Weinstein R, et al. Analysis of patient claims data to determine the prevalence of hidradenitis suppurativa in the United States. <i>J Am Acad Dermatol. </i>2013;68:412-419. <span class="citation-doi">doi:10.1016/j.jaad.2012.07.027<br/><br/></span>58. Hollestein LM, de Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989-2008. <i>Eur J Cancer. </i>2012;48:2046-2053. <span class="citation-doi">doi:10.1016/j.ejca.2012.01.003<br/><br/></span>59. Uva L, Freitas J, Soares de Almeida L, et al. Squamous cell carcinoma arising in ulcerated necrobiosis lipoidica diabeticorum. <i>Int Wound J. </i>2015;12:741-743. <span class="citation-doi">doi:10.1111/iwj.12206<br/><br/></span>60. McGrath JA, Schofield OM, Mayou BJ, et al. Epidermolysis bullosa complicated by squamous cell carcinoma: report of 10 cases. <i>J Cutan Pathol. </i>1992;19:116-123. <span class="citation-doi">doi:10.1111/j.1600-0560.1992.tb01352.x<br/><br/></span>61. <span class="authors-list-item">Montaudié</span><span class="author-sup-separator"> </span>H, Chiaverini C, Sbidian E, et al. Inherited epidermolysis bullosa and squamous cell carcinoma: a systematic review of 117 cases. <i>Orphanet J Rare Dis. </i>2016;11:117. <span class="citation-doi">doi:10.1186/s13023-016-0489-9.<br/><br/></span>62. Fine J-D. Inherited epidermolysis bullosa: past, present, and future. <i>Ann N Y Acad Sci. </i>2010;1194:213-222. <span class="citation-doi">doi:10.1111/j.1749-6632.2010.05463.x<br/><br/></span>63. Kim M, Li M, Intong-Wheeler LRA, et al. Epidemiology and outcome of squamous cell carcinoma in epidermolysis bullosa in Australia and New Zealand. <i>Acta Derm Venereol. </i>2018;98:70-76. <span class="citation-doi">doi:10.2340/00015555-2781<br/><br/></span>64. Bruckner-Tuderman L, Mitsuhashi Y, Schnyder UW, et al. Anchoring fibrils and type VII collagen are absent from skin in severe recessive dystrophic epidermolysis bullosa. <i>J Invest Dermatol. </i>1989;93:3-9. <span class="citation-doi">doi:10.1111/1523-1747.ep12277331<br/><br/></span>65. Ng Y-Z, Pourreyron C, Salas-Alanis JC, et al. Fibroblast-derived dermal matrix drives development of aggressive cutaneous squamous cell carcinoma in patients with recessive dystrophic epidermolysis bullosa. <i>Cancer Res. </i>2012;72:3522-3534. <span class="citation-doi">doi:10.1158/0008-5472.CAN-11-2996<br/><br/></span>66. Arbiser JL, Fan C-Y, Su X, et al. Involvement of p53 and p16 tumor suppressor genes in recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. <i>J Invest Dermatol. </i>2004;123:788-790. <span class="citation-doi">doi:10.1111/j.0022-202X.2004.23418.x<br/><br/></span>67. Knaup J, Gruber C, Krammer B, et al. TGFbeta-signaling in squamous cell carcinoma occurring in recessive dystrophic epidermolysis bullosa. <i>Anal Cell Pathol (Amst). </i>2011;34:339-353. <span class="citation-doi">doi:10.3233/ACP-2011-0039</span></p> <p class="reference">68. Kivisaari AK, Kallajoki M, Mirtti T, et al. Transformation-specific matrix metalloproteinases (MMP)-7 and MMP-13 are expressed by tumour cells in epidermolysis bullosa-associated squamous cell carcinomas. <i>Br J Dermatol. </i>2008;158:778-785. <span class="citation-doi">doi:10.1111/j.1365-2133.2008.08466.x<br/><br/></span>69. Rodeck U, Fertala A, Uitto J. Anchorless keratinocyte survival: an emerging pathogenic mechanism for squamous cell carcinoma in recessive dystrophic epidermolysis bullosa. <i>Exp Dermatol. </i>2007;16:465-467. <span class="citation-doi">doi:10.1111/j.1600-0625.2007.00563.x<br/><br/></span>70. Ortiz-Urda S, Garcia J, Green CL, et al. Type VII collagen is required for Ras-driven human epidermal tumorigenesis. <i>Science. </i>2005;307:1773-1776. <span class="citation-doi">doi:10.1126/science.1106209<br/><br/></span>71. Pourreyron C, Chen M, McGrath JA, et al. High levels of type VII collagen expression in recessive dystrophic epidermolysis bullosa cutaneous squamous cell carcinoma keratinocytes increases PI3K and MAPK signalling, cell migration and invasion. <i>Br J Dermatol. </i>2014;170:1256-1265. <span class="citation-doi">doi:10.1111/bjd.12715<br/><br/></span>72. Purdie KJ, Pourreyron C, Fassihi H, et al. No evidence that human papillomavirus is responsible for the aggressive nature of recessive dystrophic epidermolysis bullosa-associated squamous cell carcinoma. <i>J Invest Dermatol. </i>2010;130:2853-2855. <span class="citation-doi">doi:10.1038/jid.2010.243<br/><br/></span>73. South AP, O’Toole EA. Understanding the pathogenesis of recessive dystrophic epidermolysis bullosa squamous cell carcinoma. <i>Dermatol Clin. </i>2010;28:171-178. <span class="citation-doi">doi:10.1016/j.det.2009.10.023</span></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Department of Dermatology, Tulane University, New Orleans, Louisiana. Dr. Kuraitis also is from Roswell Park Cancer Center, Buffalo, New York.</p> <p class="disclosure">Dr. Kuraitis is a speaker and consultant for Ortho Dermatologics and a consultant for UCB. Dr. Murina is a speaker for AbbVie, Amgen, Bristol-Myers Squibb, Janssen, Pfizer, and UCB. She also is a consultant for AbbVie, Bristol-Meyers Squibb, Janssen, Novartis, Ortho Dermatologics, and UCB.<br/><br/>Correspondence: Drew Kuraitis, MD, PhD (dkuraiti@tulane.edu).<br/><br/>doi:10.12788/cutis.0914 </p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">PRACTICE<strong> POINTS</strong></p> <ul class="insidebody"> <li>Squamous cell carcinoma can develop within chronic inflammatory dermatoses.</li> <li>Orolabial discoid lupus erythematosus (DLE), oral lichen planus, and lichen sclerosus can lead to relatively rapid tumorigenesis. Squamous cell carcinoma arising in cutaneous DLE, hidradenitis suppurativa (HS), necrobiosis lipoidica, chronic wounds, and hypertrophic lichen planus tends to appear after decades of inflammation. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

PRACTICE POINTS

  • Squamous cell carcinoma can develop within chronic inflammatory dermatoses.
  • Orolabial discoid lupus erythematosus (DLE), oral lichen planus, and lichen sclerosus can lead to relatively rapid tumorigenesis. Squamous cell carcinoma arising in cutaneous DLE, hidradenitis suppurativa (HS), necrobiosis lipoidica, chronic wounds, and hypertrophic lichen planus tends to appear after decades of inflammation.
  • Be especially mindful of new orolabial DLE cases and chronic cases of HS and Marjolin ulcer because malignancies in these settings are particularly aggressive.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Teambase ID
180026A0.SIG
Disable zoom
Off

Low-Carbohydrate and Ketogenic Dietary Patterns for Type 2 Diabetes Management

Article Type
Changed
Fri, 01/12/2024 - 10:18

The prevalence of diabetes continues to increase despite advances in treatment options. In 2019, according to the Centers for Disease Control and Prevention (CDC), 37.1 million (14.7%) US adults had diabetes. Among adults aged ≥ 65 years, the prevalence is even higher at 29.2%.1 Research has also estimated that 45% of adults have evidence of prediabetes or diabetes.2 According to the Veterans Health Administration, almost 25% of enrolled veterans have diabetes.3

Background

Diabetes is associated with an increased risk of microvascular complications (eg, retinopathy, nephropathy, and neuropathy) and macrovascular complications (eg, atherosclerotic cardiovascular disease) and is one of the most common causes of morbidity and mortality in the US.4 In 2017, diabetes was estimated to cost $327 billion in the US, up from $261 billion in 2012.5 During this same period, the excess costs per person with diabetes increased from $8417 to $9601.5

Type 2 diabetes mellitus (T2DM) and its associated insulin resistance is typically considered a chronic disease with progressive loss of β-cell function. Controlling glycemia, delaying microvascular changes, and preventing macrovascular disease are major management goals. Lifestyle interventions are essential in the management and prevention of T2DM. Medication management for T2DM usually progresses through several medications, ending in insulin therapy.6 Within 10 years of diagnosis, almost half of all individuals with T2DM will require insulin to manage their glycemia.7

Bariatric surgery and nutrition approaches have been successful in reversing T2DM. Recently, there has been increased interest in nutritional approaches to place T2DM in remission, reverse the disease process, and improve insulin resistance. Contrary to popular belief, before the discovery of insulin in 1921, low-carbohydrate (LC) diets were the most common treatment for T2DM.8 With the discovery of insulin and the eventual development of low-fat dietary recommendations, LC diets were no longer favored by most clinicians.8 Low-fat diets are, by definition, also high-carbohydrate diets. By the early 1980s, low-fat diets had become the standard of care dietary recommendation, and the goal for clinicians became glycemic maintenance (with increased use of medications) rather than preventing hyperglycemia.8

With growing evidence regarding the use of LC diets for T2DM, the US Department of Veterans Affairs (VA) and US Department of Defense (DoD), the American Diabetes Association (ADA), the European Association for the Study of Diabetes (EASD), Diabetes Canada, and Diabetes Australia all include LC diets as a viable option for treating T2DM.4,9-12 This article will highlight a case using a reduced carbohydrate approach in lifestyle management and provide clinicians with practical guidance in its implementation. We will review the evidence that informs these guidelines, describe a practical approach to nutritional counseling, and review medication management and deprescribing approaches. Finally, barriers to implementation will be explored.

ILLUSTRATIVE CASE

A 64-year-old woman presented to the clinical pharmacist for the management of T2DM after her tenth hospitalization related to hyperglycemia in 10 years. She had previously been managed by primary care clinicians, clinical dietitians, endocrinologists, and certified diabetes care and education specialists. Pertinent history included diabetic ketoacidosis, coronary artery disease, hyperlipidemia, hypertension, obstructive sleep apnea, obesity, metabolic dysfunction-associated steatotic liver disease, and mild nonproliferative diabetic retinopathy with clinically significant macular edema. The patient expressed frustration with poor glycemic control during her many years of insulin therapy and an inability to lose weight due to insulin dose titrations. The patient reported prior education including but not limited to standardized sample menus, consistent carbohydrate intake, calorie reduction, general healthful nutrition, and the “move more, eat less” approach. The patient was unable to titrate insulin dosage and did not experience weight loss despite compliance with these methods.

Her medications included glargine insulin 45 units once daily, aspart insulin 5 units before meals 3 times daily, and metformin 1000 mg twice daily. Her hemoglobin A1c (HbA1c) level was 11.8%. A review of prior therapies for T2DM included glyburide 5 mg twice daily, metformin 1000 mg twice daily, 70/30 insulin (up to 340 units/d), glargine insulin (range, 10-140 units/d), regular insulin (range, 30-240 units/d), aspart insulin (range, 15-45 units/d), and U-500 regular insulin (range, 125-390 units/d). She took metoprolol 25 mg extended release daily and hydrochlorothiazide 25 mg daily, but both were discontinued after the most recent hospitalization. A review of HbA1c readings showed poor glycemic control for > 12 years (range, 10.3% to > 12.3%).

Education for lifestyle modifications, including an LC diet, was presented to the patient to assist with weight loss, improve glycemic control, and reduce insulin resistance. In addition, a glucagon-like peptide-1 agonist (liraglutide) was added to her pharmacotherapy. Continued dietary modifications with LC intake led to consistent reductions in glargine and aspart insulin therapy. The patient remained motivated throughout clinic visits due to improved glycemic control with sustainable dietary modifications, consistently reported feeling better overall, and deprescribed diabetes drug therapies. She remained off her blood pressure medications. After4 months of LC dietary modifications, all insulin therapy was discontinued. She continued with liraglutide 1.8 mg daily and metformin 1000 mg twice daily with an HbA1c of 6.3%. Two months later, her HbA1c level was 6.0%. She also lost 8 lb and her body mass index improved from 31 to 29.

 

 

Low-Carbohydrate T2DM DIET MANAGEMENT

LC diets are commonly defined as < 130 g of carbohydrates per day.13 Very LC ketogenic (VLCK) diets often contain ≤ 50 g of carbohydrates per day to induce nutritional ketosis.13 One of the first randomized controlled trials (RCTs) that compared a VLCK diet (< 30 g of carbohydrates per day) with a low-fat diet for obesity demonstrated greater weight loss at 6 months with the LC diet. In addition, patients with diabetes randomized to the LC group also showed improved insulin sensitivity. Notably, this study was done in a population of veterans enrolled at the VA Philadelphia Health Care System.14

A 2008 study comparing an LC diet with a calorie-restricted, low-glycemic diet for individuals with T2DM found that the LC diet group experienced a greater reduction in HbA1c and insulin levels and weight.15 Comparing these 2 diet groups after 24 weeks, 95% of individuals in the LC group reduced or discontinued T2DM medications vs 62% in the low-glycemic group.15 Another study of individuals with T2DM compared a VLCK diet with a low-fat diet. After 34 weeks, 55% of individuals in the LC diet group achieved an HbA1c level below the threshold for diabetes vs 0% in the low-fat diet group.16 A 2018 study of patients with T2DM investigated the impact of a very LC diet compared with the standard of care.17 After 1 year, the LC diet group experienced a mean HbA1c reduction of 1.3%, and 60% of individuals who completed the study achieved an HbA1c level < 6.5% without T2DM medications (not including metformin). This study also demonstrated that medications were significantly reduced, including 100% discontinuation of sulfonylureas and 94% reduction or elimination of insulin.

A recent study of an LC diet (< 20% energy from carbohydrates) demonstrated reduced HbA1c levels, weight, and waist circumference vs a control diet after 6 months. The control diet derived 50% to 60% of energy from carbohydrates.18 This study is typical of other LC interventions, which did not calorie restrict and instead allowed ad libitum intake.14,15

table_1.png

With mounting evidence, the VA/DoD guidelines on T2DM management included LC diets as dietary options for treating T2DM. The ADA also determined that LC diets had the most evidence in improving glycemia and included LC diets as an option for medical nutrition therapy (Table 1).10,19

A systematic review and meta-analysis looking at RCTs of LC diets found evidence for remission of T2DM without significant adverse effects (AEs).20 Another recent systematic review and network meta-analysis of 42 RCTs found that the ketogenic diet was superior for a reduction in HbA1c levels compared with 9 other dietary patterns, including low-fat, Mediterranean, and vegetarian/vegan diets. Overall, ketogenic, Mediterranean, moderate-carbohydrate, and low-glycemic index diets demonstrated improved glycemic control.21

Ideally, a comprehensive behavioral program, such as the VA Move! or Whole Health program, should incorporate patient aligned care teams (PACTs), behavioral health clinicians, clinical pharmacists, and dietitians to provide medical-nutrition therapy using LC diets. However, many facilities may not have adequate experience, expertise, or support. We provide practical approaches to provide LC nutrition counseling, medication management, and deprescribing for any primary care clinician applying LC diets for their patients. For simplicity and practicality, we define 3 types of LC dietary patterns: (1) VLCK (< 50 g); (2) LC (50-100 g); and (3) moderate LC (101-150 g).

Nutrition

table_2.png

All nutrition approaches, including LC diets, should be patient centered, individualized, and sensitive to the patient's culture. Typically, many patients have previously been instructed to consume low-fat (and subsequently) high-carbohydrate (> 150 g) meals. Most well-meaning clinicians have provided common-approach diet education from mainstream health organizations in the form of standardized handouts. For example, the Carbohydrate Counting for People with Diabetes patient education handout from the Academy of Nutrition and Dietetics provides a sample menu with 3 meals and 1 snack totaling 195 g of carbohydrates.22 In contrast, an example ADA diet has sample diets with 3 meals and 2 snacks with approximately 20 to 70 g of carbohydrates.23 In the VA, there are excellent resources to review and standardize handouts that emphasize an LC nutrition approach to T2DM, including ketogenic versions.24,25 Table 2 shows example meal plans based on different LC patterns—VLCK, LC, and moderate LC.

 

 

Starting an LC dietary pattern should maximize nutrient-dense and minimally processed proteins. Clinicians should begin with a baseline nutritional assessment through a 24-hour recall or food diary. After this has been completed, the patient’s baseline diet is assessed, and a gradual carbohydrate reduction plan is discussed. Generally, carbohydrate reduction is recommended at 1 meal per day per week. High-carbohydrate meals and snacks are restructured to favor satiating, minimally processed, high-protein food sources. Individual food preferences are considered and included in the recommended LC plan. For example, LC diets can be formulated for vegetarians and vegans as well as those who prefer meat and seafood. Prioritizing satiating and nutrient-dense foods can help increase the probability of diet acceptance and adherence.

A recent studyshowed that restricting carbohydrates at breakfast reduces 24-hour postprandial hyperglycemia and improves glycemic variability.26 Many patients consume upward of 50 g of carbohydrates at breakfast.27 For example, it is not uncommon for a patient to consume cereal with milk or oatmeal, orange juice, a banana, and toast at breakfast. Instead, the patient is advised to consume any combination of eggs, meat, no-sugar-added Greek yogurt, or berries.

To keep things simple for lunch and dinner, the patient is offered high-quality, minimally processed protein of their choosing with any nonstarchy vegetable. Should a patient desire additional carbohydrates with meals, they may reduce the baseline serving of carbohydrates by 50%. For example, if a patient normally fills 50% of their plate with spaghetti, they may reduce the pasta portion to 25% and add a meatball or increase the amount of vegetables consumed with the meal to satiety.

Snacks may include cheese, eggs, peanut butter, nuts, seeds, berries, no-sugar-added Greek yogurt, or guacamole. Oftentimes, when LC meals are adopted, the desire or need for snacking is diminished due to the satiating effect of high-quality protein sources and nonstarchy vegetables.

Adverse Effects

AEs have been reported with VLCK diets, including headache, diarrhea, constipation, muscle cramps, halitosis, light-headedness, and muscle weakness.28 These AEs may be mitigated with increased fluid intake, sodium intake, and magnesium supplementation.29 Increasing fluids to a minimum of 2 L/d and adding sodium (eg, bouillon supplementation) can minimize AEs.30 Milk of magnesia (5 mL) or slow-release magnesium chloride 200 mEq/d is suggested to reduce muscle cramps.30 There have been no studies looking at sodium intake and worsening hypertension or chronic heart failure in the setting of an LC diet, but fluid and electrolyte intake should be monitored closely, especially in patients with uncontrolled hypertension and heart failure. Other concerns of higher protein on worsening kidney function have generally not been founded.31 In some individuals, an LC and higher fat diet may increase low-density lipoprotein cholesterol (LDL-C).32 Therefore a baseline lipid panel is recommended and should be monitored along with HbA1c levels. An elevated LDL-C response may be managed by increasing protein and reducing saturated fat intake while maintaining the reduced carbohydrate content of the diet.

Medication Management

table_3.png

The adoption of an LC diet can cause a swift and profound reduction in blood sugar.33 Utilizing PACTs can help prevent adverse drug events by involving clinical pharmacists to provide recommendations and dose reductions as patients adopt an LC diet. Each approach must be individualized to the patient and can depend on several factors, including the number and strength of medications, the degree of carbohydrate reduction, baseline blood glucose, as well as assessing for medical literacy and ability to implement recommendations. Additionally, patients should monitor their blood sugar regularly and communicate with their primary care team (pharmacist, PACT registered nurse, primary care clinician, and registered dietician). Ultimately, the goal when adopting an LC diet while taking antihyperglycemics is safely avoiding hypoglycemia while reducing the number of medications the patient is taking. We summarize a practical approach to medication management that was recently published (Table 3).33,34

 

 

Medications to Reduce or Discontinue

table_4.png

Medications that can cause hypoglycemia should be the first to be reduced or discontinued upon starting an LC diet, including bolus insulin (although a small amount may be needed to correct for high blood sugar), sulfonylureas, and meglitinides. Combination insulin should be stopped and changed to basal insulin to avoid the risk of hypoglycemia (see Table 4 for insulin deprescribing recommendations). The mechanism of action in preventing the breakdown of carbohydrates in the gastrointestinal tract makes the use of α-glucosidase inhibitors superfluous, and they can be discontinued, reducing pill burden and polypharmacy risks. Sodium-glucose transport protein 2 inhibitors (SGLT2i) should be discontinued for patients on VLCK diets due to the risk of euglycemic diabetic ketoacidosis. However, with LC and moderate LC plans, the SGLT2i may be used with caution as long as patients are made aware of ketoacidosis symptoms. To help prevent the risk of hypoglycemia, basal/long-acting insulin can be continued, but at a 50% reduced dose. Patients should closely monitor blood sugar to assess for appropriateness of dose reductions. While thiazolidinediones are not contraindicated, clinicians can consider discontinuation given both their penchant for inducing weight gain and their limited outcomes data.

Medications to Continue

Medications that pose minimal risk for hypoglycemia can be continued, including metformin, dipeptidyl peptidase 4 inhibitors, and glucagon-like peptide-1 agonists. However, even though these may pose a low risk of hypoglycemia, patients should still closely monitor their blood glucose so medications can be deprescribed as soon as safely and reasonably possible.

Other Medications

The improvement in metabolic health with the reduction of carbohydrates can render other classes of medications unnecessary or require adjustment. Patients should be counseled to monitor their blood pressure as significant and rapid improvements can occur. In the event of a systolic blood pressure of 100 to 110 mm Hg or signs of hypotension, down titration or discontinuation of antihypertensives should be initiated. Limited evidence exists on the preferred order of discontinuation but should be informed by other comorbidities, such as coronary artery disease and chronic kidney disease. Given an LC diet’s diuretic effect, tapering and stopping diuretics may be an option. Other medications requiring closer monitoring include lithium (can be affected by fluid and electrolyte shifts), warfarin (may alter vitamin K intake), valproate (which may be reduced), and zonisamide and topiramate (kidney stone risk).

Remission of T2DM with LC Diets

As patients adopt LC diets and medications are deprescribed and glycemia improves, HbA1c and fasting glucose levels may drop below the diagnostic threshold for T2DM.20 As new evidence emerges surrounding the management of T2DM from a lifestyle perspective, major health care organizations have acknowledged that T2DM is not necessarily an incurable, progressive disease, but rather a disease that can be reversed or put in remission.35-37 In 2016, the World Health Organization (WHO) global report on diabetes acknowledged that T2DM reversal can be achieved via weight loss and calorie restriction.35

In 2021, a consensus statement from the ADA, the Endocrine Society, the EASD, and Diabetes UK defined T2DM remission as an HbA1c level < 6.5% for at least 3 months with no T2DM medications.36 Diabetes Australia also published a position statement in 2021 about T2DM remission.37 Like the WHO, Diabetes Australia acknowledged that remission of T2DM is possible following intensive dietary changes or bariatric surgery.37 Before the 2021 consensus statement, some experts argued that excluding metformin from the T2DM medication list may not be warranted since metformin has indications beyond T2DM. In this case, remission of T2DM could be defined as an HbA1c level < 6.5% for at least 3 months and on metformin or no T2DM medications.8  

 

 

Emerging Strategies

Emerging strategies, such as continuous glucose monitors (CGMs) and the use of intermittent fasting/time-restricted eating (TRE), can be used with the LC diet to help improve the monitoring and management of T2DM. In the recently published VA/DoD guidelines for T2DM, the work group suggested real-time CGMs for qualified patients with T2DM.4 These include patients on daily insulin who are not achieving glycemic control or to reduce the risk for hypoglycemia. CGMs have shown evidence of improved glycemic control and decreased hypoglycemia in those with T2DM.38,39 It is currently unknown if CGMs improve long-term glycemic control, but they appear promising for managing and reducing medications for those on an LC diet.40

TRE can be supplemented with an LC plan that incorporates “eating windows.” Common patterns include 14 hours of fasting and a 10-hour eating window (14F:10E), or 16 hours of fasting and an 8-hour eating window (16F:8E). By eating only in the specified window, patients generally reduce caloric intake and minimize insulin and glucose excursions during the fasting window. No changes need to be made to the macronutrient composition of the diet, and LC approaches can be used with TRE. The mechanism of action is likely multifactorial, targeting hyperinsulinemia and insulin resistance as well as producing a caloric deficit to enable weight loss.41 Eating windows may improve insulin sensitivity, reduce insulin resistance, and enhance overall glycemic control. The recent VA/DoD guidelines recommended against intermittent fasting due to concerns over the risk of hypoglycemia despite larger weight loss in TRE groups.4 Recently, a study using CGMs and TRE demonstrated both improved glycemic control and no hypoglycemic episodes in patients with T2DM on insulin.42 Patients who would like to supplement TRE with an LC plan as a strategy for improved glycemic control should work closely with their PACT to help manage their TRE and LC plan and consider a CGM adjunct, especially if on insulin.

Barriers

Managing T2DM often requires comprehensive lifestyle modifications of nutrition, exercise, sleep, stress management, and other psychosocial issues, as well as an interdisciplinary team-based approach.43 The advantage of working within the VA includes a uniform system within a network of care. However, many patients continue to use both federal and private health care. This use of out-of-network care may result in fragmented, potentially disjointed, or even contradictory dietary advice.

The VA PACT, whole health for holistic health, and weight loss interventions such as the MOVE! program provide lifestyle interventions like nutrition, physical activity, and behavior change. However, these well-intentioned approaches may provide alternative and even diverging recommendations, which place additional barriers to effective patient management. In patients who are advised and accept a trial of an LC plan, each member of the team should embrace the self-management decision of the patient and support the plan.29 Any conflicts, questions, or concerns should be communicated directly with the team in an interdisciplinary approach to provide a unified message and counsel.

The long-term effects and sustainability of an LC diet have been questioned in the literature.44-46 Recently, the use of an app-based coaching plan has demonstrated short- and long-term sustainability on an LC diet.47 In just 5 months in a large VA system, 590 patients using a virtual coaching platform and a VLCK diet plan were found to have lower HbA1c levels, reduced diabetic medication fills, lower body mass index, fewer outpatient visits, and lower prescription drug costs.

A 5-year follow-up found nearly 50% of participants sustained a VLCK diet for T2DM. For patients who participated in the study after 2 years, 72% sustained the VLCK diet in years 2 to 5. Most required nearly 50% fewer medications and in those that started with insulin, half did not require it at 5 years.48 Further research, however, is necessary to determine the long-term effects on cardiometabolic markers and health with LC diets. There are no long-term RCTs on outcomes data looking at T2DM morbidity or mortality. While there are prospective cohort studies on LC diets in the general population on mortality, they demonstrate mixed results. These studies may be confounded by heterogeneous definitions of LC diets, diet quality, and other health factors.49-51

Conclusions

The effective use of LC diets within a PACT with close and intensive lifestyle counseling and a safe approach to medication management and deprescribing can improve glycemic control, reduce the overall need for insulin, reduce medication use, and provide sustained weight loss. Additionally, the use of therapeutic carbohydrate reduction and subsequent medication deprescription may lead to sustained remission of T2DM. The current efficacy and sustainment of therapeutic carbohydrate reduction for patients with T2DM appears promising. Further research on LC diets, emerging strategies, and long-term effects on cardiometabolic risk factors, morbidity, and mortality will continue to inform future practice in our health care system.

Acknowledgments

We thank Cecile Seth who has been instrumental in pushing us forward and the Metabolic Multiplier group who has helped encourage and provide input into this article.

References

1. Centers for Disease Control and Prevention. Prevalence of Both Diagnosed and Undiagnosed Diabetes. Updated September 30, 2022. Accessed October 6, 2023. https://www.cdc.gov/diabetes/data/statistics-report/diagnosed-undiagnosed-diabetes.html

2. Centers for Disease Control and Prevention. Diabetes and Prediabetes. Updated September 6, 2022. Accessed October 6, 2023. https://www.cdc.gov/chronicdisease/resources/publications/factsheets/diabetes-prediabetes.htm 3. US Department of Veterans Affairs. Diabetes information - Nutrition and food services. Updated May 4, 2023. Accessed October 6, 2023. https://www.nutrition.va.gov/diabetes.asp

4. US Department of Veterans Affairs. Management of Type 2 Diabetes Mellitus (2023) - VA/DoD Clinical Practice Guidelines. Updated September 1, 2023. Accessed October 6, 2023. https://www.healthquality.va.gov/guidelines/CD/diabetes/

5. American Diabetes Association. Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care. 2018;41(5):917-928. doi:10.2337/dci18-0007

6. Home P, Riddle M, Cefalu WT, et al. Insulin therapy in people with type 2 diabetes: opportunities and challenges?. Diabetes Care. 2014;37(6):1499-1508. doi:10.2337/dc13-2743

7. Donath MY, Ehses JA, Maedler K, et al. Mechanisms of β-cell death in type 2 diabetes. Diabetes. 2005;54(suppl 2):S108-S113. doi:10.2337/DIABETES.54.SUPPL_2.S108

8. Hallberg SJ, Gershuni VM, Hazbun TL, Athinarayanan SJ. Reversing type 2 diabetes: a narrative review of the evidence. Nutrients. 2019;11(4):766. Published 2019 Apr 1. doi:10.3390/nu11040766

9. Davies MJ, D’Alessio DA, Fradkin J, et al. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2018;41(12):2669. doi:10.2337/DCI18-0033

10. Evert AB, Dennison M, Gardner CD, et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. Diabetes Care. 2019;42(5):731-754. doi:10.2337/DCI19-0014

11. Diabetes Canada position statement on low-carbohydrate diets for adults with diabetes: a rapid review. Can J Diabetes. 2020;44(4):295-299. doi:10.1016/J.JCJD.2020.04.001

12. Diabetes Australia. Position statements. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/research-advocacy/position-statements/

13. Feinman RD, Pogozelski WK, Astrup A, et al. Dietary carbohydrate restriction as the first approach in diabetes management: critical review and evidence base. Nutrition. 2014;31(1):1-13. doi:10.1016/j.nut.2014.06.011

14. Samaha FF, Iqbal N, Seshadri P, et al. A low-carbohydrate as compared with a low-fat diet in severe obesity. N Engl J Med. 2003;348(21):2074-2081. doi:10.1056/NEJMOA02263715. Westman EC, Yancy WS, Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutr Metab (Lond). 2008;5(1):36. doi:10.1186/1743-7075-5-36

16. Saslow LR, Mason AE, Kim S, et al. An online intervention comparing a very low-carbohydrate ketogenic diet and lifestyle recommendations versus a plate method diet in overweight individuals with type 2 diabetes: a randomized controlled trial. J Med Internet Res. 2017;19(2). doi:10.2196/JMIR.5806

17. Hallberg SJ, McKenzie AL, Williams PT, et al. Effectiveness and safety of a novel care model for the management of type 2 diabetes at 1 year: an open-label, non-randomized, controlled study. Diabetes Ther. 2018;9(2):583-612. doi:10.1007/S13300-018-0373-9

18. Gram-Kampmann EM, Hansen CD, Hugger MB, et al. Effects of a 6-month, low-carbohydrate diet on glycaemic control, body composition, and cardiovascular risk factors in patients with type 2 diabetes: An open-label randomized controlled trial. Diabetes Obes Metab. 2022;24(4):693-703. doi:10.1111/DOM.14633

19. Committee ADAPP. 5. Facilitating behavior change and well-being to improve health outcomes: standards of medical care in diabetes—2022. Diabetes Care. 2022;45(suppl 1):S60-S82. doi:10.2337/DC22-S005

20. Goldenberg JZ, Johnston BC. Low and very low carbohydrate diets for diabetes remission. BMJ. 2021;373:m4743. doi:10.1136/BMJ.N262

<--pagebreak-->

21. Jing T, Zhang S, Bai M, et al. Effect of dietary approaches on glycemic control in patients with type 2 diabetes: a systematic review with network meta-analysis of randomized trials. Nutrients. 2023;15(14):3156. doi:10.3390/nu15143156

22. Academy of Nutrition and Dietetics. Nutrition care manual. Accessed October 6, 2023. https://www.nutritioncaremanual.org/

23. Low carbohydrate and very low carbohydrate eating patterns in adults with diabetes. ShopDiabetes.org. Accessed August 5, 2022. https://shopdiabetes.org/products/low-carbohydrate-and-very-low-carbohydrate-eating-patterns-in-adults-with-diabetes-a-guide-for-health-care-providers

24. US Department of Veterans Affairs. Diabetes education - nutrition and food services. Published July 31, 2022. http://vaww.nutrition.va.gov/docs/pted/ModifiedKetogenicDiet.pdf [Source not verified]

25. US Department of Veterans Affairs, My HealtheVet. Lowdown on low-carb diets. Updated June 1, 2021. Accessed October 6, 2023. https://www.myhealth.va.gov/mhv-portal-web/ss20190724-low-carb-diet

26. Chang CR, Francois ME, Little JP. Restricting carbohydrates at breakfast is sufficient to reduce 24-hour exposure to postprandial hyperglycemia and improve glycemic variability. Am J Clin Nutr. 2019;109(5):1302-1309. doi:10.1093/AJCN/NQY261

27. Hall KD, Ayuketah A, Brychta R, et al. Ultra-processed diets cause excess calorie intake and weight gain: an inpatient randomized controlled trial of ad libitum food intake. Cell Metab. 2019;30(1):226. doi:10.1016/j.cmet.2019.05.020

28. Harvey CJ d. C, Schofield GM, Zinn C, Thornley S. Effects of differing levels of carbohydrate restriction on mood achievement of nutritional ketosis, and symptoms of carbohydrate withdrawal in healthy adults: a randomized clinical trial. Nutrition. 2019;67-68:100005. doi:10.1016/J.NUTX.2019.100005

29. Griauzde DH, Standafer Lopez K, Saslow LR, Richardson CR. A pragmatic approach to translating low- and very low-carbohydrate diets into clinical practice for patients with obesity and type 2 diabetes. Front Nutr. 2021;8:416. doi:10.3389/FNUT.2021.682137/BIBTEX

30. Westman EC, Tondt J, Maguire E, Yancy WS. Implementing a low-carbohydrate, ketogenic diet to manage type 2 diabetes mellitus. Expert Rev Endocrinol Metab. 2018;13(5):263-272. doi:10.1080/17446651.2018.1523713

31. Suyoto PST. Effect of low-carbohydrate diet on markers of renal function in patients with type 2 diabetes: a meta-analysis. Diabetes Metab Res Rev. 2018;34(7). doi:10.1002/DMRR.3032

32. Norwitz NG, Feldman D, Soto-Mota A, Kalayjian T, Ludwig DS. Elevated LDL cholesterol with a carbohydrate-restricted diet: evidence for a “lean mass hyper-responder” phenotype. Curr Dev Nutr. 2021;6(1). doi:10.1093/CDN/NZAB144

33. Murdoch C, Unwin D, Cavan D, Cucuzzella M, Patel M. Adapting diabetes medication for low carbohydrate management of type 2 diabetes: a practical guide. Br J Gen Pract. 2019;69(684):360-361. doi:10.3399/bjgp19X704525

34. Cucuzzella M, Riley K, Isaacs D. Adapting medication for type 2 diabetes to a low carbohydrate diet. Front Nutr. 2021;8:486. doi:10.3389/FNUT.2021.688540/BIBTEX

35. World Health Organization. Global report on diabetes. 2016. Accessed October 6, 2023. https://iris.who.int/bitstream/handle/10665/204871/9789241565257_eng.pdf?sequence=1

36. Riddle MC, Cefalu WT, Evans PH, et al. Consensus report: definition and interpretation of remission in type 2 diabetes. Diabetes Care. 2021;44(10):2438-2444. doi:10.2337/DCI21-0034

37. Diabetes Australia. Type 2 Diabetes remission position statement. 2021. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/wp-content/uploads/2021_Diabetes-Australia-Position-Statement_Type-2-diabetes-remission_2.pdf

38. Martens T, Beck RW, Bailey R, et al. Effect of continuous glucose monitoring on glycemic control in patients with type 2 diabetes treated with basal insulin: a randomized clinical trial. JAMA. 2021;325(22):2262-2272. doi:10.1001/JAMA.2021.7444

39. Jackson MA, Ahmann A, Shah VN. Type 2 diabetes and the use of real-time continuous glucose monitoring. Diabetes Technol Ther. 2021;23(S1):S27-S34. doi:10.1089/DIA.2021.0007

40. Oser TK, Cucuzzella M, Stasinopoulos M, Moncrief M, McCall A, Cox DJ. An innovative, paradigm-shifting lifestyle intervention to reduce glucose excursions with the use of continuous glucose monitoring to educate, motivate, and activate adults with newly diagnosed type 2 diabetes: pilot feasibility study. JMIR Diabetes. 2022;7(1). doi:10.2196/34465

41. Światkiewicz I, Woźniak A, Taub PR. Time-restricted eating and metabolic syndrome: current status and future perspectives. Nutrients. 2021;13(1):221. doi:10.3390/NU13010221

42. Obermayer A, Tripolt NJ, Pferschy PN, et al. Efficacy and safety of intermittent fasting in people with insulin-treated type 2 diabetes (INTERFAST-2)—a randomized controlled trial. Diabetes Care. 2023;46(2):463-468. doi:10.2337/dc22-1622

43. American Diabetes Association. 5. Lifestyle management: standards of medical care in diabetes—2019. Diabetes Care. 2019;42(suppl 1):S46-S60. doi:10.2337/DC19-S005

44. Li S, Ding L, Xiao X. Comparing the efficacy and safety of low-carbohydrate diets with low-fat diets for type 2 diabetes mellitus patients: a systematic review and meta-analysis of randomized clinical trials. Int J Endocrinol. 2021;2021:8521756. Published 2021 Dec 6. doi:10.1155/2021/8521756

45. Choi JH, Kang JH, Chon S. Comprehensive understanding for application in Korean patients with type 2 diabetes mellitus of the consensus statement on carbohydrate-restricted diets by Korean Diabetes Association, Korean Society for the Study of Obesity, and Korean Society of Hypertension. Diabetes Metab J. 2022;46(3):377. doi:10.4093/DMJ.2022.0051

46. Jayedi A, Zeraattalab-Motlagh S, Jabbarzadeh B, et al. Dose-dependent effect of carbohydrate restriction for type 2 diabetes management: a systematic review and dose-response meta-analysis of randomized controlled trials. Am J Clin Nutr. 2022;116(1). doi:10.1093/AJCN/NQAC066

47. Strombotne KL, Lum J, Ndugga NJ, et al. Effectiveness of a ketogenic diet and virtual coaching intervention for patients with diabetes: a difference-in-differences analysis. Diabetes Obes Metab. 2021;23(12):2643-2650. doi:10.1111/DOM.14515

48. Virta Health. Virta Health highlights lasting, transformative health improvements in 5-year diabetes reversal study. June 5, 2022. Accessed October 6, 2023. https://www.virtahealth.com/blog/virta-sustainable-health-improvements-5-year-diabetes-reversal-study

49. Wan Z, Shan Z, Geng T, et al. Associations of moderate low-carbohydrate diets with mortality among patients with type 2 diabetes: a prospective cohort study. J Clin Endocrinol Metab. 2022;107(7):E2702-E2709. doi:10.1210/CLINEM/DGAC235

50. Akter S, Mizoue T, Nanri A, et al. Low carbohydrate diet and all cause and cause-specific mortality. Clin Nutr. 2021;40(4):2016-2024. doi:10.1016/J.CLNU.2020.09.022

51. Shan Z, Guo Y, Hu FB, Liu L, Qi Q. Association of low-carbohydrate and low-fat diets with mortality among US adults. JAMA Intern Med. 2020;180(4):513-523. doi:10.1001/JAMAINTERNMED.2019.6980

Article PDF
Author and Disclosure Information

Robert C. Oh, MD, MPHa; Kendrick C. Murphy, PharmD, BCACP, MHPb; Cory M. Jenks, PharmD, MHP, BCPS, BCACPc;  Kathleen B. Lopez, RDN, CDCES, CNSCd; Mahendra A. Patel, PharmD, BCPSe; Emily E. Scotland, MSN, FNP-Ce;  Monu Khanna, MD, MHPf

Correspondence:  Robert Oh (robert.oh@va.gov)

aVeterans Affairs Palo Alto Health Care System, California

bWestern North Carolina Veterans Affairs Health Care System, Asheville

cAmbulatory Care Clinical Pharmacist Society of Metabolic Health Practitioners, Tucson, Arizona

dVeterans Affairs Boston Health Care System, Massachusetts

eSouthern Arizona Veterans Affairs Health Care System, Tucson

fVeterans Affairs St Louis Health Care System, Missouri

Author disclosures
CM Jenks is married to an employee of Virta Medical, which provides care related to type 2 diabetes and ketogenic diets.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent
Written consent for publication has been obtained from the patient reported in the illustrative case.

Issue
Federal Practitioner - 41(1)a
Publications
Topics
Page Number
6
Sections
Author and Disclosure Information

Robert C. Oh, MD, MPHa; Kendrick C. Murphy, PharmD, BCACP, MHPb; Cory M. Jenks, PharmD, MHP, BCPS, BCACPc;  Kathleen B. Lopez, RDN, CDCES, CNSCd; Mahendra A. Patel, PharmD, BCPSe; Emily E. Scotland, MSN, FNP-Ce;  Monu Khanna, MD, MHPf

Correspondence:  Robert Oh (robert.oh@va.gov)

aVeterans Affairs Palo Alto Health Care System, California

bWestern North Carolina Veterans Affairs Health Care System, Asheville

cAmbulatory Care Clinical Pharmacist Society of Metabolic Health Practitioners, Tucson, Arizona

dVeterans Affairs Boston Health Care System, Massachusetts

eSouthern Arizona Veterans Affairs Health Care System, Tucson

fVeterans Affairs St Louis Health Care System, Missouri

Author disclosures
CM Jenks is married to an employee of Virta Medical, which provides care related to type 2 diabetes and ketogenic diets.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent
Written consent for publication has been obtained from the patient reported in the illustrative case.

Author and Disclosure Information

Robert C. Oh, MD, MPHa; Kendrick C. Murphy, PharmD, BCACP, MHPb; Cory M. Jenks, PharmD, MHP, BCPS, BCACPc;  Kathleen B. Lopez, RDN, CDCES, CNSCd; Mahendra A. Patel, PharmD, BCPSe; Emily E. Scotland, MSN, FNP-Ce;  Monu Khanna, MD, MHPf

Correspondence:  Robert Oh (robert.oh@va.gov)

aVeterans Affairs Palo Alto Health Care System, California

bWestern North Carolina Veterans Affairs Health Care System, Asheville

cAmbulatory Care Clinical Pharmacist Society of Metabolic Health Practitioners, Tucson, Arizona

dVeterans Affairs Boston Health Care System, Massachusetts

eSouthern Arizona Veterans Affairs Health Care System, Tucson

fVeterans Affairs St Louis Health Care System, Missouri

Author disclosures
CM Jenks is married to an employee of Virta Medical, which provides care related to type 2 diabetes and ketogenic diets.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent
Written consent for publication has been obtained from the patient reported in the illustrative case.

Article PDF
Article PDF

The prevalence of diabetes continues to increase despite advances in treatment options. In 2019, according to the Centers for Disease Control and Prevention (CDC), 37.1 million (14.7%) US adults had diabetes. Among adults aged ≥ 65 years, the prevalence is even higher at 29.2%.1 Research has also estimated that 45% of adults have evidence of prediabetes or diabetes.2 According to the Veterans Health Administration, almost 25% of enrolled veterans have diabetes.3

Background

Diabetes is associated with an increased risk of microvascular complications (eg, retinopathy, nephropathy, and neuropathy) and macrovascular complications (eg, atherosclerotic cardiovascular disease) and is one of the most common causes of morbidity and mortality in the US.4 In 2017, diabetes was estimated to cost $327 billion in the US, up from $261 billion in 2012.5 During this same period, the excess costs per person with diabetes increased from $8417 to $9601.5

Type 2 diabetes mellitus (T2DM) and its associated insulin resistance is typically considered a chronic disease with progressive loss of β-cell function. Controlling glycemia, delaying microvascular changes, and preventing macrovascular disease are major management goals. Lifestyle interventions are essential in the management and prevention of T2DM. Medication management for T2DM usually progresses through several medications, ending in insulin therapy.6 Within 10 years of diagnosis, almost half of all individuals with T2DM will require insulin to manage their glycemia.7

Bariatric surgery and nutrition approaches have been successful in reversing T2DM. Recently, there has been increased interest in nutritional approaches to place T2DM in remission, reverse the disease process, and improve insulin resistance. Contrary to popular belief, before the discovery of insulin in 1921, low-carbohydrate (LC) diets were the most common treatment for T2DM.8 With the discovery of insulin and the eventual development of low-fat dietary recommendations, LC diets were no longer favored by most clinicians.8 Low-fat diets are, by definition, also high-carbohydrate diets. By the early 1980s, low-fat diets had become the standard of care dietary recommendation, and the goal for clinicians became glycemic maintenance (with increased use of medications) rather than preventing hyperglycemia.8

With growing evidence regarding the use of LC diets for T2DM, the US Department of Veterans Affairs (VA) and US Department of Defense (DoD), the American Diabetes Association (ADA), the European Association for the Study of Diabetes (EASD), Diabetes Canada, and Diabetes Australia all include LC diets as a viable option for treating T2DM.4,9-12 This article will highlight a case using a reduced carbohydrate approach in lifestyle management and provide clinicians with practical guidance in its implementation. We will review the evidence that informs these guidelines, describe a practical approach to nutritional counseling, and review medication management and deprescribing approaches. Finally, barriers to implementation will be explored.

ILLUSTRATIVE CASE

A 64-year-old woman presented to the clinical pharmacist for the management of T2DM after her tenth hospitalization related to hyperglycemia in 10 years. She had previously been managed by primary care clinicians, clinical dietitians, endocrinologists, and certified diabetes care and education specialists. Pertinent history included diabetic ketoacidosis, coronary artery disease, hyperlipidemia, hypertension, obstructive sleep apnea, obesity, metabolic dysfunction-associated steatotic liver disease, and mild nonproliferative diabetic retinopathy with clinically significant macular edema. The patient expressed frustration with poor glycemic control during her many years of insulin therapy and an inability to lose weight due to insulin dose titrations. The patient reported prior education including but not limited to standardized sample menus, consistent carbohydrate intake, calorie reduction, general healthful nutrition, and the “move more, eat less” approach. The patient was unable to titrate insulin dosage and did not experience weight loss despite compliance with these methods.

Her medications included glargine insulin 45 units once daily, aspart insulin 5 units before meals 3 times daily, and metformin 1000 mg twice daily. Her hemoglobin A1c (HbA1c) level was 11.8%. A review of prior therapies for T2DM included glyburide 5 mg twice daily, metformin 1000 mg twice daily, 70/30 insulin (up to 340 units/d), glargine insulin (range, 10-140 units/d), regular insulin (range, 30-240 units/d), aspart insulin (range, 15-45 units/d), and U-500 regular insulin (range, 125-390 units/d). She took metoprolol 25 mg extended release daily and hydrochlorothiazide 25 mg daily, but both were discontinued after the most recent hospitalization. A review of HbA1c readings showed poor glycemic control for > 12 years (range, 10.3% to > 12.3%).

Education for lifestyle modifications, including an LC diet, was presented to the patient to assist with weight loss, improve glycemic control, and reduce insulin resistance. In addition, a glucagon-like peptide-1 agonist (liraglutide) was added to her pharmacotherapy. Continued dietary modifications with LC intake led to consistent reductions in glargine and aspart insulin therapy. The patient remained motivated throughout clinic visits due to improved glycemic control with sustainable dietary modifications, consistently reported feeling better overall, and deprescribed diabetes drug therapies. She remained off her blood pressure medications. After4 months of LC dietary modifications, all insulin therapy was discontinued. She continued with liraglutide 1.8 mg daily and metformin 1000 mg twice daily with an HbA1c of 6.3%. Two months later, her HbA1c level was 6.0%. She also lost 8 lb and her body mass index improved from 31 to 29.

 

 

Low-Carbohydrate T2DM DIET MANAGEMENT

LC diets are commonly defined as < 130 g of carbohydrates per day.13 Very LC ketogenic (VLCK) diets often contain ≤ 50 g of carbohydrates per day to induce nutritional ketosis.13 One of the first randomized controlled trials (RCTs) that compared a VLCK diet (< 30 g of carbohydrates per day) with a low-fat diet for obesity demonstrated greater weight loss at 6 months with the LC diet. In addition, patients with diabetes randomized to the LC group also showed improved insulin sensitivity. Notably, this study was done in a population of veterans enrolled at the VA Philadelphia Health Care System.14

A 2008 study comparing an LC diet with a calorie-restricted, low-glycemic diet for individuals with T2DM found that the LC diet group experienced a greater reduction in HbA1c and insulin levels and weight.15 Comparing these 2 diet groups after 24 weeks, 95% of individuals in the LC group reduced or discontinued T2DM medications vs 62% in the low-glycemic group.15 Another study of individuals with T2DM compared a VLCK diet with a low-fat diet. After 34 weeks, 55% of individuals in the LC diet group achieved an HbA1c level below the threshold for diabetes vs 0% in the low-fat diet group.16 A 2018 study of patients with T2DM investigated the impact of a very LC diet compared with the standard of care.17 After 1 year, the LC diet group experienced a mean HbA1c reduction of 1.3%, and 60% of individuals who completed the study achieved an HbA1c level < 6.5% without T2DM medications (not including metformin). This study also demonstrated that medications were significantly reduced, including 100% discontinuation of sulfonylureas and 94% reduction or elimination of insulin.

A recent study of an LC diet (< 20% energy from carbohydrates) demonstrated reduced HbA1c levels, weight, and waist circumference vs a control diet after 6 months. The control diet derived 50% to 60% of energy from carbohydrates.18 This study is typical of other LC interventions, which did not calorie restrict and instead allowed ad libitum intake.14,15

table_1.png

With mounting evidence, the VA/DoD guidelines on T2DM management included LC diets as dietary options for treating T2DM. The ADA also determined that LC diets had the most evidence in improving glycemia and included LC diets as an option for medical nutrition therapy (Table 1).10,19

A systematic review and meta-analysis looking at RCTs of LC diets found evidence for remission of T2DM without significant adverse effects (AEs).20 Another recent systematic review and network meta-analysis of 42 RCTs found that the ketogenic diet was superior for a reduction in HbA1c levels compared with 9 other dietary patterns, including low-fat, Mediterranean, and vegetarian/vegan diets. Overall, ketogenic, Mediterranean, moderate-carbohydrate, and low-glycemic index diets demonstrated improved glycemic control.21

Ideally, a comprehensive behavioral program, such as the VA Move! or Whole Health program, should incorporate patient aligned care teams (PACTs), behavioral health clinicians, clinical pharmacists, and dietitians to provide medical-nutrition therapy using LC diets. However, many facilities may not have adequate experience, expertise, or support. We provide practical approaches to provide LC nutrition counseling, medication management, and deprescribing for any primary care clinician applying LC diets for their patients. For simplicity and practicality, we define 3 types of LC dietary patterns: (1) VLCK (< 50 g); (2) LC (50-100 g); and (3) moderate LC (101-150 g).

Nutrition

table_2.png

All nutrition approaches, including LC diets, should be patient centered, individualized, and sensitive to the patient's culture. Typically, many patients have previously been instructed to consume low-fat (and subsequently) high-carbohydrate (> 150 g) meals. Most well-meaning clinicians have provided common-approach diet education from mainstream health organizations in the form of standardized handouts. For example, the Carbohydrate Counting for People with Diabetes patient education handout from the Academy of Nutrition and Dietetics provides a sample menu with 3 meals and 1 snack totaling 195 g of carbohydrates.22 In contrast, an example ADA diet has sample diets with 3 meals and 2 snacks with approximately 20 to 70 g of carbohydrates.23 In the VA, there are excellent resources to review and standardize handouts that emphasize an LC nutrition approach to T2DM, including ketogenic versions.24,25 Table 2 shows example meal plans based on different LC patterns—VLCK, LC, and moderate LC.

 

 

Starting an LC dietary pattern should maximize nutrient-dense and minimally processed proteins. Clinicians should begin with a baseline nutritional assessment through a 24-hour recall or food diary. After this has been completed, the patient’s baseline diet is assessed, and a gradual carbohydrate reduction plan is discussed. Generally, carbohydrate reduction is recommended at 1 meal per day per week. High-carbohydrate meals and snacks are restructured to favor satiating, minimally processed, high-protein food sources. Individual food preferences are considered and included in the recommended LC plan. For example, LC diets can be formulated for vegetarians and vegans as well as those who prefer meat and seafood. Prioritizing satiating and nutrient-dense foods can help increase the probability of diet acceptance and adherence.

A recent studyshowed that restricting carbohydrates at breakfast reduces 24-hour postprandial hyperglycemia and improves glycemic variability.26 Many patients consume upward of 50 g of carbohydrates at breakfast.27 For example, it is not uncommon for a patient to consume cereal with milk or oatmeal, orange juice, a banana, and toast at breakfast. Instead, the patient is advised to consume any combination of eggs, meat, no-sugar-added Greek yogurt, or berries.

To keep things simple for lunch and dinner, the patient is offered high-quality, minimally processed protein of their choosing with any nonstarchy vegetable. Should a patient desire additional carbohydrates with meals, they may reduce the baseline serving of carbohydrates by 50%. For example, if a patient normally fills 50% of their plate with spaghetti, they may reduce the pasta portion to 25% and add a meatball or increase the amount of vegetables consumed with the meal to satiety.

Snacks may include cheese, eggs, peanut butter, nuts, seeds, berries, no-sugar-added Greek yogurt, or guacamole. Oftentimes, when LC meals are adopted, the desire or need for snacking is diminished due to the satiating effect of high-quality protein sources and nonstarchy vegetables.

Adverse Effects

AEs have been reported with VLCK diets, including headache, diarrhea, constipation, muscle cramps, halitosis, light-headedness, and muscle weakness.28 These AEs may be mitigated with increased fluid intake, sodium intake, and magnesium supplementation.29 Increasing fluids to a minimum of 2 L/d and adding sodium (eg, bouillon supplementation) can minimize AEs.30 Milk of magnesia (5 mL) or slow-release magnesium chloride 200 mEq/d is suggested to reduce muscle cramps.30 There have been no studies looking at sodium intake and worsening hypertension or chronic heart failure in the setting of an LC diet, but fluid and electrolyte intake should be monitored closely, especially in patients with uncontrolled hypertension and heart failure. Other concerns of higher protein on worsening kidney function have generally not been founded.31 In some individuals, an LC and higher fat diet may increase low-density lipoprotein cholesterol (LDL-C).32 Therefore a baseline lipid panel is recommended and should be monitored along with HbA1c levels. An elevated LDL-C response may be managed by increasing protein and reducing saturated fat intake while maintaining the reduced carbohydrate content of the diet.

Medication Management

table_3.png

The adoption of an LC diet can cause a swift and profound reduction in blood sugar.33 Utilizing PACTs can help prevent adverse drug events by involving clinical pharmacists to provide recommendations and dose reductions as patients adopt an LC diet. Each approach must be individualized to the patient and can depend on several factors, including the number and strength of medications, the degree of carbohydrate reduction, baseline blood glucose, as well as assessing for medical literacy and ability to implement recommendations. Additionally, patients should monitor their blood sugar regularly and communicate with their primary care team (pharmacist, PACT registered nurse, primary care clinician, and registered dietician). Ultimately, the goal when adopting an LC diet while taking antihyperglycemics is safely avoiding hypoglycemia while reducing the number of medications the patient is taking. We summarize a practical approach to medication management that was recently published (Table 3).33,34

 

 

Medications to Reduce or Discontinue

table_4.png

Medications that can cause hypoglycemia should be the first to be reduced or discontinued upon starting an LC diet, including bolus insulin (although a small amount may be needed to correct for high blood sugar), sulfonylureas, and meglitinides. Combination insulin should be stopped and changed to basal insulin to avoid the risk of hypoglycemia (see Table 4 for insulin deprescribing recommendations). The mechanism of action in preventing the breakdown of carbohydrates in the gastrointestinal tract makes the use of α-glucosidase inhibitors superfluous, and they can be discontinued, reducing pill burden and polypharmacy risks. Sodium-glucose transport protein 2 inhibitors (SGLT2i) should be discontinued for patients on VLCK diets due to the risk of euglycemic diabetic ketoacidosis. However, with LC and moderate LC plans, the SGLT2i may be used with caution as long as patients are made aware of ketoacidosis symptoms. To help prevent the risk of hypoglycemia, basal/long-acting insulin can be continued, but at a 50% reduced dose. Patients should closely monitor blood sugar to assess for appropriateness of dose reductions. While thiazolidinediones are not contraindicated, clinicians can consider discontinuation given both their penchant for inducing weight gain and their limited outcomes data.

Medications to Continue

Medications that pose minimal risk for hypoglycemia can be continued, including metformin, dipeptidyl peptidase 4 inhibitors, and glucagon-like peptide-1 agonists. However, even though these may pose a low risk of hypoglycemia, patients should still closely monitor their blood glucose so medications can be deprescribed as soon as safely and reasonably possible.

Other Medications

The improvement in metabolic health with the reduction of carbohydrates can render other classes of medications unnecessary or require adjustment. Patients should be counseled to monitor their blood pressure as significant and rapid improvements can occur. In the event of a systolic blood pressure of 100 to 110 mm Hg or signs of hypotension, down titration or discontinuation of antihypertensives should be initiated. Limited evidence exists on the preferred order of discontinuation but should be informed by other comorbidities, such as coronary artery disease and chronic kidney disease. Given an LC diet’s diuretic effect, tapering and stopping diuretics may be an option. Other medications requiring closer monitoring include lithium (can be affected by fluid and electrolyte shifts), warfarin (may alter vitamin K intake), valproate (which may be reduced), and zonisamide and topiramate (kidney stone risk).

Remission of T2DM with LC Diets

As patients adopt LC diets and medications are deprescribed and glycemia improves, HbA1c and fasting glucose levels may drop below the diagnostic threshold for T2DM.20 As new evidence emerges surrounding the management of T2DM from a lifestyle perspective, major health care organizations have acknowledged that T2DM is not necessarily an incurable, progressive disease, but rather a disease that can be reversed or put in remission.35-37 In 2016, the World Health Organization (WHO) global report on diabetes acknowledged that T2DM reversal can be achieved via weight loss and calorie restriction.35

In 2021, a consensus statement from the ADA, the Endocrine Society, the EASD, and Diabetes UK defined T2DM remission as an HbA1c level < 6.5% for at least 3 months with no T2DM medications.36 Diabetes Australia also published a position statement in 2021 about T2DM remission.37 Like the WHO, Diabetes Australia acknowledged that remission of T2DM is possible following intensive dietary changes or bariatric surgery.37 Before the 2021 consensus statement, some experts argued that excluding metformin from the T2DM medication list may not be warranted since metformin has indications beyond T2DM. In this case, remission of T2DM could be defined as an HbA1c level < 6.5% for at least 3 months and on metformin or no T2DM medications.8  

 

 

Emerging Strategies

Emerging strategies, such as continuous glucose monitors (CGMs) and the use of intermittent fasting/time-restricted eating (TRE), can be used with the LC diet to help improve the monitoring and management of T2DM. In the recently published VA/DoD guidelines for T2DM, the work group suggested real-time CGMs for qualified patients with T2DM.4 These include patients on daily insulin who are not achieving glycemic control or to reduce the risk for hypoglycemia. CGMs have shown evidence of improved glycemic control and decreased hypoglycemia in those with T2DM.38,39 It is currently unknown if CGMs improve long-term glycemic control, but they appear promising for managing and reducing medications for those on an LC diet.40

TRE can be supplemented with an LC plan that incorporates “eating windows.” Common patterns include 14 hours of fasting and a 10-hour eating window (14F:10E), or 16 hours of fasting and an 8-hour eating window (16F:8E). By eating only in the specified window, patients generally reduce caloric intake and minimize insulin and glucose excursions during the fasting window. No changes need to be made to the macronutrient composition of the diet, and LC approaches can be used with TRE. The mechanism of action is likely multifactorial, targeting hyperinsulinemia and insulin resistance as well as producing a caloric deficit to enable weight loss.41 Eating windows may improve insulin sensitivity, reduce insulin resistance, and enhance overall glycemic control. The recent VA/DoD guidelines recommended against intermittent fasting due to concerns over the risk of hypoglycemia despite larger weight loss in TRE groups.4 Recently, a study using CGMs and TRE demonstrated both improved glycemic control and no hypoglycemic episodes in patients with T2DM on insulin.42 Patients who would like to supplement TRE with an LC plan as a strategy for improved glycemic control should work closely with their PACT to help manage their TRE and LC plan and consider a CGM adjunct, especially if on insulin.

Barriers

Managing T2DM often requires comprehensive lifestyle modifications of nutrition, exercise, sleep, stress management, and other psychosocial issues, as well as an interdisciplinary team-based approach.43 The advantage of working within the VA includes a uniform system within a network of care. However, many patients continue to use both federal and private health care. This use of out-of-network care may result in fragmented, potentially disjointed, or even contradictory dietary advice.

The VA PACT, whole health for holistic health, and weight loss interventions such as the MOVE! program provide lifestyle interventions like nutrition, physical activity, and behavior change. However, these well-intentioned approaches may provide alternative and even diverging recommendations, which place additional barriers to effective patient management. In patients who are advised and accept a trial of an LC plan, each member of the team should embrace the self-management decision of the patient and support the plan.29 Any conflicts, questions, or concerns should be communicated directly with the team in an interdisciplinary approach to provide a unified message and counsel.

The long-term effects and sustainability of an LC diet have been questioned in the literature.44-46 Recently, the use of an app-based coaching plan has demonstrated short- and long-term sustainability on an LC diet.47 In just 5 months in a large VA system, 590 patients using a virtual coaching platform and a VLCK diet plan were found to have lower HbA1c levels, reduced diabetic medication fills, lower body mass index, fewer outpatient visits, and lower prescription drug costs.

A 5-year follow-up found nearly 50% of participants sustained a VLCK diet for T2DM. For patients who participated in the study after 2 years, 72% sustained the VLCK diet in years 2 to 5. Most required nearly 50% fewer medications and in those that started with insulin, half did not require it at 5 years.48 Further research, however, is necessary to determine the long-term effects on cardiometabolic markers and health with LC diets. There are no long-term RCTs on outcomes data looking at T2DM morbidity or mortality. While there are prospective cohort studies on LC diets in the general population on mortality, they demonstrate mixed results. These studies may be confounded by heterogeneous definitions of LC diets, diet quality, and other health factors.49-51

Conclusions

The effective use of LC diets within a PACT with close and intensive lifestyle counseling and a safe approach to medication management and deprescribing can improve glycemic control, reduce the overall need for insulin, reduce medication use, and provide sustained weight loss. Additionally, the use of therapeutic carbohydrate reduction and subsequent medication deprescription may lead to sustained remission of T2DM. The current efficacy and sustainment of therapeutic carbohydrate reduction for patients with T2DM appears promising. Further research on LC diets, emerging strategies, and long-term effects on cardiometabolic risk factors, morbidity, and mortality will continue to inform future practice in our health care system.

Acknowledgments

We thank Cecile Seth who has been instrumental in pushing us forward and the Metabolic Multiplier group who has helped encourage and provide input into this article.

The prevalence of diabetes continues to increase despite advances in treatment options. In 2019, according to the Centers for Disease Control and Prevention (CDC), 37.1 million (14.7%) US adults had diabetes. Among adults aged ≥ 65 years, the prevalence is even higher at 29.2%.1 Research has also estimated that 45% of adults have evidence of prediabetes or diabetes.2 According to the Veterans Health Administration, almost 25% of enrolled veterans have diabetes.3

Background

Diabetes is associated with an increased risk of microvascular complications (eg, retinopathy, nephropathy, and neuropathy) and macrovascular complications (eg, atherosclerotic cardiovascular disease) and is one of the most common causes of morbidity and mortality in the US.4 In 2017, diabetes was estimated to cost $327 billion in the US, up from $261 billion in 2012.5 During this same period, the excess costs per person with diabetes increased from $8417 to $9601.5

Type 2 diabetes mellitus (T2DM) and its associated insulin resistance is typically considered a chronic disease with progressive loss of β-cell function. Controlling glycemia, delaying microvascular changes, and preventing macrovascular disease are major management goals. Lifestyle interventions are essential in the management and prevention of T2DM. Medication management for T2DM usually progresses through several medications, ending in insulin therapy.6 Within 10 years of diagnosis, almost half of all individuals with T2DM will require insulin to manage their glycemia.7

Bariatric surgery and nutrition approaches have been successful in reversing T2DM. Recently, there has been increased interest in nutritional approaches to place T2DM in remission, reverse the disease process, and improve insulin resistance. Contrary to popular belief, before the discovery of insulin in 1921, low-carbohydrate (LC) diets were the most common treatment for T2DM.8 With the discovery of insulin and the eventual development of low-fat dietary recommendations, LC diets were no longer favored by most clinicians.8 Low-fat diets are, by definition, also high-carbohydrate diets. By the early 1980s, low-fat diets had become the standard of care dietary recommendation, and the goal for clinicians became glycemic maintenance (with increased use of medications) rather than preventing hyperglycemia.8

With growing evidence regarding the use of LC diets for T2DM, the US Department of Veterans Affairs (VA) and US Department of Defense (DoD), the American Diabetes Association (ADA), the European Association for the Study of Diabetes (EASD), Diabetes Canada, and Diabetes Australia all include LC diets as a viable option for treating T2DM.4,9-12 This article will highlight a case using a reduced carbohydrate approach in lifestyle management and provide clinicians with practical guidance in its implementation. We will review the evidence that informs these guidelines, describe a practical approach to nutritional counseling, and review medication management and deprescribing approaches. Finally, barriers to implementation will be explored.

ILLUSTRATIVE CASE

A 64-year-old woman presented to the clinical pharmacist for the management of T2DM after her tenth hospitalization related to hyperglycemia in 10 years. She had previously been managed by primary care clinicians, clinical dietitians, endocrinologists, and certified diabetes care and education specialists. Pertinent history included diabetic ketoacidosis, coronary artery disease, hyperlipidemia, hypertension, obstructive sleep apnea, obesity, metabolic dysfunction-associated steatotic liver disease, and mild nonproliferative diabetic retinopathy with clinically significant macular edema. The patient expressed frustration with poor glycemic control during her many years of insulin therapy and an inability to lose weight due to insulin dose titrations. The patient reported prior education including but not limited to standardized sample menus, consistent carbohydrate intake, calorie reduction, general healthful nutrition, and the “move more, eat less” approach. The patient was unable to titrate insulin dosage and did not experience weight loss despite compliance with these methods.

Her medications included glargine insulin 45 units once daily, aspart insulin 5 units before meals 3 times daily, and metformin 1000 mg twice daily. Her hemoglobin A1c (HbA1c) level was 11.8%. A review of prior therapies for T2DM included glyburide 5 mg twice daily, metformin 1000 mg twice daily, 70/30 insulin (up to 340 units/d), glargine insulin (range, 10-140 units/d), regular insulin (range, 30-240 units/d), aspart insulin (range, 15-45 units/d), and U-500 regular insulin (range, 125-390 units/d). She took metoprolol 25 mg extended release daily and hydrochlorothiazide 25 mg daily, but both were discontinued after the most recent hospitalization. A review of HbA1c readings showed poor glycemic control for > 12 years (range, 10.3% to > 12.3%).

Education for lifestyle modifications, including an LC diet, was presented to the patient to assist with weight loss, improve glycemic control, and reduce insulin resistance. In addition, a glucagon-like peptide-1 agonist (liraglutide) was added to her pharmacotherapy. Continued dietary modifications with LC intake led to consistent reductions in glargine and aspart insulin therapy. The patient remained motivated throughout clinic visits due to improved glycemic control with sustainable dietary modifications, consistently reported feeling better overall, and deprescribed diabetes drug therapies. She remained off her blood pressure medications. After4 months of LC dietary modifications, all insulin therapy was discontinued. She continued with liraglutide 1.8 mg daily and metformin 1000 mg twice daily with an HbA1c of 6.3%. Two months later, her HbA1c level was 6.0%. She also lost 8 lb and her body mass index improved from 31 to 29.

 

 

Low-Carbohydrate T2DM DIET MANAGEMENT

LC diets are commonly defined as < 130 g of carbohydrates per day.13 Very LC ketogenic (VLCK) diets often contain ≤ 50 g of carbohydrates per day to induce nutritional ketosis.13 One of the first randomized controlled trials (RCTs) that compared a VLCK diet (< 30 g of carbohydrates per day) with a low-fat diet for obesity demonstrated greater weight loss at 6 months with the LC diet. In addition, patients with diabetes randomized to the LC group also showed improved insulin sensitivity. Notably, this study was done in a population of veterans enrolled at the VA Philadelphia Health Care System.14

A 2008 study comparing an LC diet with a calorie-restricted, low-glycemic diet for individuals with T2DM found that the LC diet group experienced a greater reduction in HbA1c and insulin levels and weight.15 Comparing these 2 diet groups after 24 weeks, 95% of individuals in the LC group reduced or discontinued T2DM medications vs 62% in the low-glycemic group.15 Another study of individuals with T2DM compared a VLCK diet with a low-fat diet. After 34 weeks, 55% of individuals in the LC diet group achieved an HbA1c level below the threshold for diabetes vs 0% in the low-fat diet group.16 A 2018 study of patients with T2DM investigated the impact of a very LC diet compared with the standard of care.17 After 1 year, the LC diet group experienced a mean HbA1c reduction of 1.3%, and 60% of individuals who completed the study achieved an HbA1c level < 6.5% without T2DM medications (not including metformin). This study also demonstrated that medications were significantly reduced, including 100% discontinuation of sulfonylureas and 94% reduction or elimination of insulin.

A recent study of an LC diet (< 20% energy from carbohydrates) demonstrated reduced HbA1c levels, weight, and waist circumference vs a control diet after 6 months. The control diet derived 50% to 60% of energy from carbohydrates.18 This study is typical of other LC interventions, which did not calorie restrict and instead allowed ad libitum intake.14,15

table_1.png

With mounting evidence, the VA/DoD guidelines on T2DM management included LC diets as dietary options for treating T2DM. The ADA also determined that LC diets had the most evidence in improving glycemia and included LC diets as an option for medical nutrition therapy (Table 1).10,19

A systematic review and meta-analysis looking at RCTs of LC diets found evidence for remission of T2DM without significant adverse effects (AEs).20 Another recent systematic review and network meta-analysis of 42 RCTs found that the ketogenic diet was superior for a reduction in HbA1c levels compared with 9 other dietary patterns, including low-fat, Mediterranean, and vegetarian/vegan diets. Overall, ketogenic, Mediterranean, moderate-carbohydrate, and low-glycemic index diets demonstrated improved glycemic control.21

Ideally, a comprehensive behavioral program, such as the VA Move! or Whole Health program, should incorporate patient aligned care teams (PACTs), behavioral health clinicians, clinical pharmacists, and dietitians to provide medical-nutrition therapy using LC diets. However, many facilities may not have adequate experience, expertise, or support. We provide practical approaches to provide LC nutrition counseling, medication management, and deprescribing for any primary care clinician applying LC diets for their patients. For simplicity and practicality, we define 3 types of LC dietary patterns: (1) VLCK (< 50 g); (2) LC (50-100 g); and (3) moderate LC (101-150 g).

Nutrition

table_2.png

All nutrition approaches, including LC diets, should be patient centered, individualized, and sensitive to the patient's culture. Typically, many patients have previously been instructed to consume low-fat (and subsequently) high-carbohydrate (> 150 g) meals. Most well-meaning clinicians have provided common-approach diet education from mainstream health organizations in the form of standardized handouts. For example, the Carbohydrate Counting for People with Diabetes patient education handout from the Academy of Nutrition and Dietetics provides a sample menu with 3 meals and 1 snack totaling 195 g of carbohydrates.22 In contrast, an example ADA diet has sample diets with 3 meals and 2 snacks with approximately 20 to 70 g of carbohydrates.23 In the VA, there are excellent resources to review and standardize handouts that emphasize an LC nutrition approach to T2DM, including ketogenic versions.24,25 Table 2 shows example meal plans based on different LC patterns—VLCK, LC, and moderate LC.

 

 

Starting an LC dietary pattern should maximize nutrient-dense and minimally processed proteins. Clinicians should begin with a baseline nutritional assessment through a 24-hour recall or food diary. After this has been completed, the patient’s baseline diet is assessed, and a gradual carbohydrate reduction plan is discussed. Generally, carbohydrate reduction is recommended at 1 meal per day per week. High-carbohydrate meals and snacks are restructured to favor satiating, minimally processed, high-protein food sources. Individual food preferences are considered and included in the recommended LC plan. For example, LC diets can be formulated for vegetarians and vegans as well as those who prefer meat and seafood. Prioritizing satiating and nutrient-dense foods can help increase the probability of diet acceptance and adherence.

A recent studyshowed that restricting carbohydrates at breakfast reduces 24-hour postprandial hyperglycemia and improves glycemic variability.26 Many patients consume upward of 50 g of carbohydrates at breakfast.27 For example, it is not uncommon for a patient to consume cereal with milk or oatmeal, orange juice, a banana, and toast at breakfast. Instead, the patient is advised to consume any combination of eggs, meat, no-sugar-added Greek yogurt, or berries.

To keep things simple for lunch and dinner, the patient is offered high-quality, minimally processed protein of their choosing with any nonstarchy vegetable. Should a patient desire additional carbohydrates with meals, they may reduce the baseline serving of carbohydrates by 50%. For example, if a patient normally fills 50% of their plate with spaghetti, they may reduce the pasta portion to 25% and add a meatball or increase the amount of vegetables consumed with the meal to satiety.

Snacks may include cheese, eggs, peanut butter, nuts, seeds, berries, no-sugar-added Greek yogurt, or guacamole. Oftentimes, when LC meals are adopted, the desire or need for snacking is diminished due to the satiating effect of high-quality protein sources and nonstarchy vegetables.

Adverse Effects

AEs have been reported with VLCK diets, including headache, diarrhea, constipation, muscle cramps, halitosis, light-headedness, and muscle weakness.28 These AEs may be mitigated with increased fluid intake, sodium intake, and magnesium supplementation.29 Increasing fluids to a minimum of 2 L/d and adding sodium (eg, bouillon supplementation) can minimize AEs.30 Milk of magnesia (5 mL) or slow-release magnesium chloride 200 mEq/d is suggested to reduce muscle cramps.30 There have been no studies looking at sodium intake and worsening hypertension or chronic heart failure in the setting of an LC diet, but fluid and electrolyte intake should be monitored closely, especially in patients with uncontrolled hypertension and heart failure. Other concerns of higher protein on worsening kidney function have generally not been founded.31 In some individuals, an LC and higher fat diet may increase low-density lipoprotein cholesterol (LDL-C).32 Therefore a baseline lipid panel is recommended and should be monitored along with HbA1c levels. An elevated LDL-C response may be managed by increasing protein and reducing saturated fat intake while maintaining the reduced carbohydrate content of the diet.

Medication Management

table_3.png

The adoption of an LC diet can cause a swift and profound reduction in blood sugar.33 Utilizing PACTs can help prevent adverse drug events by involving clinical pharmacists to provide recommendations and dose reductions as patients adopt an LC diet. Each approach must be individualized to the patient and can depend on several factors, including the number and strength of medications, the degree of carbohydrate reduction, baseline blood glucose, as well as assessing for medical literacy and ability to implement recommendations. Additionally, patients should monitor their blood sugar regularly and communicate with their primary care team (pharmacist, PACT registered nurse, primary care clinician, and registered dietician). Ultimately, the goal when adopting an LC diet while taking antihyperglycemics is safely avoiding hypoglycemia while reducing the number of medications the patient is taking. We summarize a practical approach to medication management that was recently published (Table 3).33,34

 

 

Medications to Reduce or Discontinue

table_4.png

Medications that can cause hypoglycemia should be the first to be reduced or discontinued upon starting an LC diet, including bolus insulin (although a small amount may be needed to correct for high blood sugar), sulfonylureas, and meglitinides. Combination insulin should be stopped and changed to basal insulin to avoid the risk of hypoglycemia (see Table 4 for insulin deprescribing recommendations). The mechanism of action in preventing the breakdown of carbohydrates in the gastrointestinal tract makes the use of α-glucosidase inhibitors superfluous, and they can be discontinued, reducing pill burden and polypharmacy risks. Sodium-glucose transport protein 2 inhibitors (SGLT2i) should be discontinued for patients on VLCK diets due to the risk of euglycemic diabetic ketoacidosis. However, with LC and moderate LC plans, the SGLT2i may be used with caution as long as patients are made aware of ketoacidosis symptoms. To help prevent the risk of hypoglycemia, basal/long-acting insulin can be continued, but at a 50% reduced dose. Patients should closely monitor blood sugar to assess for appropriateness of dose reductions. While thiazolidinediones are not contraindicated, clinicians can consider discontinuation given both their penchant for inducing weight gain and their limited outcomes data.

Medications to Continue

Medications that pose minimal risk for hypoglycemia can be continued, including metformin, dipeptidyl peptidase 4 inhibitors, and glucagon-like peptide-1 agonists. However, even though these may pose a low risk of hypoglycemia, patients should still closely monitor their blood glucose so medications can be deprescribed as soon as safely and reasonably possible.

Other Medications

The improvement in metabolic health with the reduction of carbohydrates can render other classes of medications unnecessary or require adjustment. Patients should be counseled to monitor their blood pressure as significant and rapid improvements can occur. In the event of a systolic blood pressure of 100 to 110 mm Hg or signs of hypotension, down titration or discontinuation of antihypertensives should be initiated. Limited evidence exists on the preferred order of discontinuation but should be informed by other comorbidities, such as coronary artery disease and chronic kidney disease. Given an LC diet’s diuretic effect, tapering and stopping diuretics may be an option. Other medications requiring closer monitoring include lithium (can be affected by fluid and electrolyte shifts), warfarin (may alter vitamin K intake), valproate (which may be reduced), and zonisamide and topiramate (kidney stone risk).

Remission of T2DM with LC Diets

As patients adopt LC diets and medications are deprescribed and glycemia improves, HbA1c and fasting glucose levels may drop below the diagnostic threshold for T2DM.20 As new evidence emerges surrounding the management of T2DM from a lifestyle perspective, major health care organizations have acknowledged that T2DM is not necessarily an incurable, progressive disease, but rather a disease that can be reversed or put in remission.35-37 In 2016, the World Health Organization (WHO) global report on diabetes acknowledged that T2DM reversal can be achieved via weight loss and calorie restriction.35

In 2021, a consensus statement from the ADA, the Endocrine Society, the EASD, and Diabetes UK defined T2DM remission as an HbA1c level < 6.5% for at least 3 months with no T2DM medications.36 Diabetes Australia also published a position statement in 2021 about T2DM remission.37 Like the WHO, Diabetes Australia acknowledged that remission of T2DM is possible following intensive dietary changes or bariatric surgery.37 Before the 2021 consensus statement, some experts argued that excluding metformin from the T2DM medication list may not be warranted since metformin has indications beyond T2DM. In this case, remission of T2DM could be defined as an HbA1c level < 6.5% for at least 3 months and on metformin or no T2DM medications.8  

 

 

Emerging Strategies

Emerging strategies, such as continuous glucose monitors (CGMs) and the use of intermittent fasting/time-restricted eating (TRE), can be used with the LC diet to help improve the monitoring and management of T2DM. In the recently published VA/DoD guidelines for T2DM, the work group suggested real-time CGMs for qualified patients with T2DM.4 These include patients on daily insulin who are not achieving glycemic control or to reduce the risk for hypoglycemia. CGMs have shown evidence of improved glycemic control and decreased hypoglycemia in those with T2DM.38,39 It is currently unknown if CGMs improve long-term glycemic control, but they appear promising for managing and reducing medications for those on an LC diet.40

TRE can be supplemented with an LC plan that incorporates “eating windows.” Common patterns include 14 hours of fasting and a 10-hour eating window (14F:10E), or 16 hours of fasting and an 8-hour eating window (16F:8E). By eating only in the specified window, patients generally reduce caloric intake and minimize insulin and glucose excursions during the fasting window. No changes need to be made to the macronutrient composition of the diet, and LC approaches can be used with TRE. The mechanism of action is likely multifactorial, targeting hyperinsulinemia and insulin resistance as well as producing a caloric deficit to enable weight loss.41 Eating windows may improve insulin sensitivity, reduce insulin resistance, and enhance overall glycemic control. The recent VA/DoD guidelines recommended against intermittent fasting due to concerns over the risk of hypoglycemia despite larger weight loss in TRE groups.4 Recently, a study using CGMs and TRE demonstrated both improved glycemic control and no hypoglycemic episodes in patients with T2DM on insulin.42 Patients who would like to supplement TRE with an LC plan as a strategy for improved glycemic control should work closely with their PACT to help manage their TRE and LC plan and consider a CGM adjunct, especially if on insulin.

Barriers

Managing T2DM often requires comprehensive lifestyle modifications of nutrition, exercise, sleep, stress management, and other psychosocial issues, as well as an interdisciplinary team-based approach.43 The advantage of working within the VA includes a uniform system within a network of care. However, many patients continue to use both federal and private health care. This use of out-of-network care may result in fragmented, potentially disjointed, or even contradictory dietary advice.

The VA PACT, whole health for holistic health, and weight loss interventions such as the MOVE! program provide lifestyle interventions like nutrition, physical activity, and behavior change. However, these well-intentioned approaches may provide alternative and even diverging recommendations, which place additional barriers to effective patient management. In patients who are advised and accept a trial of an LC plan, each member of the team should embrace the self-management decision of the patient and support the plan.29 Any conflicts, questions, or concerns should be communicated directly with the team in an interdisciplinary approach to provide a unified message and counsel.

The long-term effects and sustainability of an LC diet have been questioned in the literature.44-46 Recently, the use of an app-based coaching plan has demonstrated short- and long-term sustainability on an LC diet.47 In just 5 months in a large VA system, 590 patients using a virtual coaching platform and a VLCK diet plan were found to have lower HbA1c levels, reduced diabetic medication fills, lower body mass index, fewer outpatient visits, and lower prescription drug costs.

A 5-year follow-up found nearly 50% of participants sustained a VLCK diet for T2DM. For patients who participated in the study after 2 years, 72% sustained the VLCK diet in years 2 to 5. Most required nearly 50% fewer medications and in those that started with insulin, half did not require it at 5 years.48 Further research, however, is necessary to determine the long-term effects on cardiometabolic markers and health with LC diets. There are no long-term RCTs on outcomes data looking at T2DM morbidity or mortality. While there are prospective cohort studies on LC diets in the general population on mortality, they demonstrate mixed results. These studies may be confounded by heterogeneous definitions of LC diets, diet quality, and other health factors.49-51

Conclusions

The effective use of LC diets within a PACT with close and intensive lifestyle counseling and a safe approach to medication management and deprescribing can improve glycemic control, reduce the overall need for insulin, reduce medication use, and provide sustained weight loss. Additionally, the use of therapeutic carbohydrate reduction and subsequent medication deprescription may lead to sustained remission of T2DM. The current efficacy and sustainment of therapeutic carbohydrate reduction for patients with T2DM appears promising. Further research on LC diets, emerging strategies, and long-term effects on cardiometabolic risk factors, morbidity, and mortality will continue to inform future practice in our health care system.

Acknowledgments

We thank Cecile Seth who has been instrumental in pushing us forward and the Metabolic Multiplier group who has helped encourage and provide input into this article.

References

1. Centers for Disease Control and Prevention. Prevalence of Both Diagnosed and Undiagnosed Diabetes. Updated September 30, 2022. Accessed October 6, 2023. https://www.cdc.gov/diabetes/data/statistics-report/diagnosed-undiagnosed-diabetes.html

2. Centers for Disease Control and Prevention. Diabetes and Prediabetes. Updated September 6, 2022. Accessed October 6, 2023. https://www.cdc.gov/chronicdisease/resources/publications/factsheets/diabetes-prediabetes.htm 3. US Department of Veterans Affairs. Diabetes information - Nutrition and food services. Updated May 4, 2023. Accessed October 6, 2023. https://www.nutrition.va.gov/diabetes.asp

4. US Department of Veterans Affairs. Management of Type 2 Diabetes Mellitus (2023) - VA/DoD Clinical Practice Guidelines. Updated September 1, 2023. Accessed October 6, 2023. https://www.healthquality.va.gov/guidelines/CD/diabetes/

5. American Diabetes Association. Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care. 2018;41(5):917-928. doi:10.2337/dci18-0007

6. Home P, Riddle M, Cefalu WT, et al. Insulin therapy in people with type 2 diabetes: opportunities and challenges?. Diabetes Care. 2014;37(6):1499-1508. doi:10.2337/dc13-2743

7. Donath MY, Ehses JA, Maedler K, et al. Mechanisms of β-cell death in type 2 diabetes. Diabetes. 2005;54(suppl 2):S108-S113. doi:10.2337/DIABETES.54.SUPPL_2.S108

8. Hallberg SJ, Gershuni VM, Hazbun TL, Athinarayanan SJ. Reversing type 2 diabetes: a narrative review of the evidence. Nutrients. 2019;11(4):766. Published 2019 Apr 1. doi:10.3390/nu11040766

9. Davies MJ, D’Alessio DA, Fradkin J, et al. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2018;41(12):2669. doi:10.2337/DCI18-0033

10. Evert AB, Dennison M, Gardner CD, et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. Diabetes Care. 2019;42(5):731-754. doi:10.2337/DCI19-0014

11. Diabetes Canada position statement on low-carbohydrate diets for adults with diabetes: a rapid review. Can J Diabetes. 2020;44(4):295-299. doi:10.1016/J.JCJD.2020.04.001

12. Diabetes Australia. Position statements. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/research-advocacy/position-statements/

13. Feinman RD, Pogozelski WK, Astrup A, et al. Dietary carbohydrate restriction as the first approach in diabetes management: critical review and evidence base. Nutrition. 2014;31(1):1-13. doi:10.1016/j.nut.2014.06.011

14. Samaha FF, Iqbal N, Seshadri P, et al. A low-carbohydrate as compared with a low-fat diet in severe obesity. N Engl J Med. 2003;348(21):2074-2081. doi:10.1056/NEJMOA02263715. Westman EC, Yancy WS, Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutr Metab (Lond). 2008;5(1):36. doi:10.1186/1743-7075-5-36

16. Saslow LR, Mason AE, Kim S, et al. An online intervention comparing a very low-carbohydrate ketogenic diet and lifestyle recommendations versus a plate method diet in overweight individuals with type 2 diabetes: a randomized controlled trial. J Med Internet Res. 2017;19(2). doi:10.2196/JMIR.5806

17. Hallberg SJ, McKenzie AL, Williams PT, et al. Effectiveness and safety of a novel care model for the management of type 2 diabetes at 1 year: an open-label, non-randomized, controlled study. Diabetes Ther. 2018;9(2):583-612. doi:10.1007/S13300-018-0373-9

18. Gram-Kampmann EM, Hansen CD, Hugger MB, et al. Effects of a 6-month, low-carbohydrate diet on glycaemic control, body composition, and cardiovascular risk factors in patients with type 2 diabetes: An open-label randomized controlled trial. Diabetes Obes Metab. 2022;24(4):693-703. doi:10.1111/DOM.14633

19. Committee ADAPP. 5. Facilitating behavior change and well-being to improve health outcomes: standards of medical care in diabetes—2022. Diabetes Care. 2022;45(suppl 1):S60-S82. doi:10.2337/DC22-S005

20. Goldenberg JZ, Johnston BC. Low and very low carbohydrate diets for diabetes remission. BMJ. 2021;373:m4743. doi:10.1136/BMJ.N262

<--pagebreak-->

21. Jing T, Zhang S, Bai M, et al. Effect of dietary approaches on glycemic control in patients with type 2 diabetes: a systematic review with network meta-analysis of randomized trials. Nutrients. 2023;15(14):3156. doi:10.3390/nu15143156

22. Academy of Nutrition and Dietetics. Nutrition care manual. Accessed October 6, 2023. https://www.nutritioncaremanual.org/

23. Low carbohydrate and very low carbohydrate eating patterns in adults with diabetes. ShopDiabetes.org. Accessed August 5, 2022. https://shopdiabetes.org/products/low-carbohydrate-and-very-low-carbohydrate-eating-patterns-in-adults-with-diabetes-a-guide-for-health-care-providers

24. US Department of Veterans Affairs. Diabetes education - nutrition and food services. Published July 31, 2022. http://vaww.nutrition.va.gov/docs/pted/ModifiedKetogenicDiet.pdf [Source not verified]

25. US Department of Veterans Affairs, My HealtheVet. Lowdown on low-carb diets. Updated June 1, 2021. Accessed October 6, 2023. https://www.myhealth.va.gov/mhv-portal-web/ss20190724-low-carb-diet

26. Chang CR, Francois ME, Little JP. Restricting carbohydrates at breakfast is sufficient to reduce 24-hour exposure to postprandial hyperglycemia and improve glycemic variability. Am J Clin Nutr. 2019;109(5):1302-1309. doi:10.1093/AJCN/NQY261

27. Hall KD, Ayuketah A, Brychta R, et al. Ultra-processed diets cause excess calorie intake and weight gain: an inpatient randomized controlled trial of ad libitum food intake. Cell Metab. 2019;30(1):226. doi:10.1016/j.cmet.2019.05.020

28. Harvey CJ d. C, Schofield GM, Zinn C, Thornley S. Effects of differing levels of carbohydrate restriction on mood achievement of nutritional ketosis, and symptoms of carbohydrate withdrawal in healthy adults: a randomized clinical trial. Nutrition. 2019;67-68:100005. doi:10.1016/J.NUTX.2019.100005

29. Griauzde DH, Standafer Lopez K, Saslow LR, Richardson CR. A pragmatic approach to translating low- and very low-carbohydrate diets into clinical practice for patients with obesity and type 2 diabetes. Front Nutr. 2021;8:416. doi:10.3389/FNUT.2021.682137/BIBTEX

30. Westman EC, Tondt J, Maguire E, Yancy WS. Implementing a low-carbohydrate, ketogenic diet to manage type 2 diabetes mellitus. Expert Rev Endocrinol Metab. 2018;13(5):263-272. doi:10.1080/17446651.2018.1523713

31. Suyoto PST. Effect of low-carbohydrate diet on markers of renal function in patients with type 2 diabetes: a meta-analysis. Diabetes Metab Res Rev. 2018;34(7). doi:10.1002/DMRR.3032

32. Norwitz NG, Feldman D, Soto-Mota A, Kalayjian T, Ludwig DS. Elevated LDL cholesterol with a carbohydrate-restricted diet: evidence for a “lean mass hyper-responder” phenotype. Curr Dev Nutr. 2021;6(1). doi:10.1093/CDN/NZAB144

33. Murdoch C, Unwin D, Cavan D, Cucuzzella M, Patel M. Adapting diabetes medication for low carbohydrate management of type 2 diabetes: a practical guide. Br J Gen Pract. 2019;69(684):360-361. doi:10.3399/bjgp19X704525

34. Cucuzzella M, Riley K, Isaacs D. Adapting medication for type 2 diabetes to a low carbohydrate diet. Front Nutr. 2021;8:486. doi:10.3389/FNUT.2021.688540/BIBTEX

35. World Health Organization. Global report on diabetes. 2016. Accessed October 6, 2023. https://iris.who.int/bitstream/handle/10665/204871/9789241565257_eng.pdf?sequence=1

36. Riddle MC, Cefalu WT, Evans PH, et al. Consensus report: definition and interpretation of remission in type 2 diabetes. Diabetes Care. 2021;44(10):2438-2444. doi:10.2337/DCI21-0034

37. Diabetes Australia. Type 2 Diabetes remission position statement. 2021. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/wp-content/uploads/2021_Diabetes-Australia-Position-Statement_Type-2-diabetes-remission_2.pdf

38. Martens T, Beck RW, Bailey R, et al. Effect of continuous glucose monitoring on glycemic control in patients with type 2 diabetes treated with basal insulin: a randomized clinical trial. JAMA. 2021;325(22):2262-2272. doi:10.1001/JAMA.2021.7444

39. Jackson MA, Ahmann A, Shah VN. Type 2 diabetes and the use of real-time continuous glucose monitoring. Diabetes Technol Ther. 2021;23(S1):S27-S34. doi:10.1089/DIA.2021.0007

40. Oser TK, Cucuzzella M, Stasinopoulos M, Moncrief M, McCall A, Cox DJ. An innovative, paradigm-shifting lifestyle intervention to reduce glucose excursions with the use of continuous glucose monitoring to educate, motivate, and activate adults with newly diagnosed type 2 diabetes: pilot feasibility study. JMIR Diabetes. 2022;7(1). doi:10.2196/34465

41. Światkiewicz I, Woźniak A, Taub PR. Time-restricted eating and metabolic syndrome: current status and future perspectives. Nutrients. 2021;13(1):221. doi:10.3390/NU13010221

42. Obermayer A, Tripolt NJ, Pferschy PN, et al. Efficacy and safety of intermittent fasting in people with insulin-treated type 2 diabetes (INTERFAST-2)—a randomized controlled trial. Diabetes Care. 2023;46(2):463-468. doi:10.2337/dc22-1622

43. American Diabetes Association. 5. Lifestyle management: standards of medical care in diabetes—2019. Diabetes Care. 2019;42(suppl 1):S46-S60. doi:10.2337/DC19-S005

44. Li S, Ding L, Xiao X. Comparing the efficacy and safety of low-carbohydrate diets with low-fat diets for type 2 diabetes mellitus patients: a systematic review and meta-analysis of randomized clinical trials. Int J Endocrinol. 2021;2021:8521756. Published 2021 Dec 6. doi:10.1155/2021/8521756

45. Choi JH, Kang JH, Chon S. Comprehensive understanding for application in Korean patients with type 2 diabetes mellitus of the consensus statement on carbohydrate-restricted diets by Korean Diabetes Association, Korean Society for the Study of Obesity, and Korean Society of Hypertension. Diabetes Metab J. 2022;46(3):377. doi:10.4093/DMJ.2022.0051

46. Jayedi A, Zeraattalab-Motlagh S, Jabbarzadeh B, et al. Dose-dependent effect of carbohydrate restriction for type 2 diabetes management: a systematic review and dose-response meta-analysis of randomized controlled trials. Am J Clin Nutr. 2022;116(1). doi:10.1093/AJCN/NQAC066

47. Strombotne KL, Lum J, Ndugga NJ, et al. Effectiveness of a ketogenic diet and virtual coaching intervention for patients with diabetes: a difference-in-differences analysis. Diabetes Obes Metab. 2021;23(12):2643-2650. doi:10.1111/DOM.14515

48. Virta Health. Virta Health highlights lasting, transformative health improvements in 5-year diabetes reversal study. June 5, 2022. Accessed October 6, 2023. https://www.virtahealth.com/blog/virta-sustainable-health-improvements-5-year-diabetes-reversal-study

49. Wan Z, Shan Z, Geng T, et al. Associations of moderate low-carbohydrate diets with mortality among patients with type 2 diabetes: a prospective cohort study. J Clin Endocrinol Metab. 2022;107(7):E2702-E2709. doi:10.1210/CLINEM/DGAC235

50. Akter S, Mizoue T, Nanri A, et al. Low carbohydrate diet and all cause and cause-specific mortality. Clin Nutr. 2021;40(4):2016-2024. doi:10.1016/J.CLNU.2020.09.022

51. Shan Z, Guo Y, Hu FB, Liu L, Qi Q. Association of low-carbohydrate and low-fat diets with mortality among US adults. JAMA Intern Med. 2020;180(4):513-523. doi:10.1001/JAMAINTERNMED.2019.6980

References

1. Centers for Disease Control and Prevention. Prevalence of Both Diagnosed and Undiagnosed Diabetes. Updated September 30, 2022. Accessed October 6, 2023. https://www.cdc.gov/diabetes/data/statistics-report/diagnosed-undiagnosed-diabetes.html

2. Centers for Disease Control and Prevention. Diabetes and Prediabetes. Updated September 6, 2022. Accessed October 6, 2023. https://www.cdc.gov/chronicdisease/resources/publications/factsheets/diabetes-prediabetes.htm 3. US Department of Veterans Affairs. Diabetes information - Nutrition and food services. Updated May 4, 2023. Accessed October 6, 2023. https://www.nutrition.va.gov/diabetes.asp

4. US Department of Veterans Affairs. Management of Type 2 Diabetes Mellitus (2023) - VA/DoD Clinical Practice Guidelines. Updated September 1, 2023. Accessed October 6, 2023. https://www.healthquality.va.gov/guidelines/CD/diabetes/

5. American Diabetes Association. Economic Costs of Diabetes in the U.S. in 2017. Diabetes Care. 2018;41(5):917-928. doi:10.2337/dci18-0007

6. Home P, Riddle M, Cefalu WT, et al. Insulin therapy in people with type 2 diabetes: opportunities and challenges?. Diabetes Care. 2014;37(6):1499-1508. doi:10.2337/dc13-2743

7. Donath MY, Ehses JA, Maedler K, et al. Mechanisms of β-cell death in type 2 diabetes. Diabetes. 2005;54(suppl 2):S108-S113. doi:10.2337/DIABETES.54.SUPPL_2.S108

8. Hallberg SJ, Gershuni VM, Hazbun TL, Athinarayanan SJ. Reversing type 2 diabetes: a narrative review of the evidence. Nutrients. 2019;11(4):766. Published 2019 Apr 1. doi:10.3390/nu11040766

9. Davies MJ, D’Alessio DA, Fradkin J, et al. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2018;41(12):2669. doi:10.2337/DCI18-0033

10. Evert AB, Dennison M, Gardner CD, et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. Diabetes Care. 2019;42(5):731-754. doi:10.2337/DCI19-0014

11. Diabetes Canada position statement on low-carbohydrate diets for adults with diabetes: a rapid review. Can J Diabetes. 2020;44(4):295-299. doi:10.1016/J.JCJD.2020.04.001

12. Diabetes Australia. Position statements. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/research-advocacy/position-statements/

13. Feinman RD, Pogozelski WK, Astrup A, et al. Dietary carbohydrate restriction as the first approach in diabetes management: critical review and evidence base. Nutrition. 2014;31(1):1-13. doi:10.1016/j.nut.2014.06.011

14. Samaha FF, Iqbal N, Seshadri P, et al. A low-carbohydrate as compared with a low-fat diet in severe obesity. N Engl J Med. 2003;348(21):2074-2081. doi:10.1056/NEJMOA02263715. Westman EC, Yancy WS, Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. Nutr Metab (Lond). 2008;5(1):36. doi:10.1186/1743-7075-5-36

16. Saslow LR, Mason AE, Kim S, et al. An online intervention comparing a very low-carbohydrate ketogenic diet and lifestyle recommendations versus a plate method diet in overweight individuals with type 2 diabetes: a randomized controlled trial. J Med Internet Res. 2017;19(2). doi:10.2196/JMIR.5806

17. Hallberg SJ, McKenzie AL, Williams PT, et al. Effectiveness and safety of a novel care model for the management of type 2 diabetes at 1 year: an open-label, non-randomized, controlled study. Diabetes Ther. 2018;9(2):583-612. doi:10.1007/S13300-018-0373-9

18. Gram-Kampmann EM, Hansen CD, Hugger MB, et al. Effects of a 6-month, low-carbohydrate diet on glycaemic control, body composition, and cardiovascular risk factors in patients with type 2 diabetes: An open-label randomized controlled trial. Diabetes Obes Metab. 2022;24(4):693-703. doi:10.1111/DOM.14633

19. Committee ADAPP. 5. Facilitating behavior change and well-being to improve health outcomes: standards of medical care in diabetes—2022. Diabetes Care. 2022;45(suppl 1):S60-S82. doi:10.2337/DC22-S005

20. Goldenberg JZ, Johnston BC. Low and very low carbohydrate diets for diabetes remission. BMJ. 2021;373:m4743. doi:10.1136/BMJ.N262

<--pagebreak-->

21. Jing T, Zhang S, Bai M, et al. Effect of dietary approaches on glycemic control in patients with type 2 diabetes: a systematic review with network meta-analysis of randomized trials. Nutrients. 2023;15(14):3156. doi:10.3390/nu15143156

22. Academy of Nutrition and Dietetics. Nutrition care manual. Accessed October 6, 2023. https://www.nutritioncaremanual.org/

23. Low carbohydrate and very low carbohydrate eating patterns in adults with diabetes. ShopDiabetes.org. Accessed August 5, 2022. https://shopdiabetes.org/products/low-carbohydrate-and-very-low-carbohydrate-eating-patterns-in-adults-with-diabetes-a-guide-for-health-care-providers

24. US Department of Veterans Affairs. Diabetes education - nutrition and food services. Published July 31, 2022. http://vaww.nutrition.va.gov/docs/pted/ModifiedKetogenicDiet.pdf [Source not verified]

25. US Department of Veterans Affairs, My HealtheVet. Lowdown on low-carb diets. Updated June 1, 2021. Accessed October 6, 2023. https://www.myhealth.va.gov/mhv-portal-web/ss20190724-low-carb-diet

26. Chang CR, Francois ME, Little JP. Restricting carbohydrates at breakfast is sufficient to reduce 24-hour exposure to postprandial hyperglycemia and improve glycemic variability. Am J Clin Nutr. 2019;109(5):1302-1309. doi:10.1093/AJCN/NQY261

27. Hall KD, Ayuketah A, Brychta R, et al. Ultra-processed diets cause excess calorie intake and weight gain: an inpatient randomized controlled trial of ad libitum food intake. Cell Metab. 2019;30(1):226. doi:10.1016/j.cmet.2019.05.020

28. Harvey CJ d. C, Schofield GM, Zinn C, Thornley S. Effects of differing levels of carbohydrate restriction on mood achievement of nutritional ketosis, and symptoms of carbohydrate withdrawal in healthy adults: a randomized clinical trial. Nutrition. 2019;67-68:100005. doi:10.1016/J.NUTX.2019.100005

29. Griauzde DH, Standafer Lopez K, Saslow LR, Richardson CR. A pragmatic approach to translating low- and very low-carbohydrate diets into clinical practice for patients with obesity and type 2 diabetes. Front Nutr. 2021;8:416. doi:10.3389/FNUT.2021.682137/BIBTEX

30. Westman EC, Tondt J, Maguire E, Yancy WS. Implementing a low-carbohydrate, ketogenic diet to manage type 2 diabetes mellitus. Expert Rev Endocrinol Metab. 2018;13(5):263-272. doi:10.1080/17446651.2018.1523713

31. Suyoto PST. Effect of low-carbohydrate diet on markers of renal function in patients with type 2 diabetes: a meta-analysis. Diabetes Metab Res Rev. 2018;34(7). doi:10.1002/DMRR.3032

32. Norwitz NG, Feldman D, Soto-Mota A, Kalayjian T, Ludwig DS. Elevated LDL cholesterol with a carbohydrate-restricted diet: evidence for a “lean mass hyper-responder” phenotype. Curr Dev Nutr. 2021;6(1). doi:10.1093/CDN/NZAB144

33. Murdoch C, Unwin D, Cavan D, Cucuzzella M, Patel M. Adapting diabetes medication for low carbohydrate management of type 2 diabetes: a practical guide. Br J Gen Pract. 2019;69(684):360-361. doi:10.3399/bjgp19X704525

34. Cucuzzella M, Riley K, Isaacs D. Adapting medication for type 2 diabetes to a low carbohydrate diet. Front Nutr. 2021;8:486. doi:10.3389/FNUT.2021.688540/BIBTEX

35. World Health Organization. Global report on diabetes. 2016. Accessed October 6, 2023. https://iris.who.int/bitstream/handle/10665/204871/9789241565257_eng.pdf?sequence=1

36. Riddle MC, Cefalu WT, Evans PH, et al. Consensus report: definition and interpretation of remission in type 2 diabetes. Diabetes Care. 2021;44(10):2438-2444. doi:10.2337/DCI21-0034

37. Diabetes Australia. Type 2 Diabetes remission position statement. 2021. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/wp-content/uploads/2021_Diabetes-Australia-Position-Statement_Type-2-diabetes-remission_2.pdf

38. Martens T, Beck RW, Bailey R, et al. Effect of continuous glucose monitoring on glycemic control in patients with type 2 diabetes treated with basal insulin: a randomized clinical trial. JAMA. 2021;325(22):2262-2272. doi:10.1001/JAMA.2021.7444

39. Jackson MA, Ahmann A, Shah VN. Type 2 diabetes and the use of real-time continuous glucose monitoring. Diabetes Technol Ther. 2021;23(S1):S27-S34. doi:10.1089/DIA.2021.0007

40. Oser TK, Cucuzzella M, Stasinopoulos M, Moncrief M, McCall A, Cox DJ. An innovative, paradigm-shifting lifestyle intervention to reduce glucose excursions with the use of continuous glucose monitoring to educate, motivate, and activate adults with newly diagnosed type 2 diabetes: pilot feasibility study. JMIR Diabetes. 2022;7(1). doi:10.2196/34465

41. Światkiewicz I, Woźniak A, Taub PR. Time-restricted eating and metabolic syndrome: current status and future perspectives. Nutrients. 2021;13(1):221. doi:10.3390/NU13010221

42. Obermayer A, Tripolt NJ, Pferschy PN, et al. Efficacy and safety of intermittent fasting in people with insulin-treated type 2 diabetes (INTERFAST-2)—a randomized controlled trial. Diabetes Care. 2023;46(2):463-468. doi:10.2337/dc22-1622

43. American Diabetes Association. 5. Lifestyle management: standards of medical care in diabetes—2019. Diabetes Care. 2019;42(suppl 1):S46-S60. doi:10.2337/DC19-S005

44. Li S, Ding L, Xiao X. Comparing the efficacy and safety of low-carbohydrate diets with low-fat diets for type 2 diabetes mellitus patients: a systematic review and meta-analysis of randomized clinical trials. Int J Endocrinol. 2021;2021:8521756. Published 2021 Dec 6. doi:10.1155/2021/8521756

45. Choi JH, Kang JH, Chon S. Comprehensive understanding for application in Korean patients with type 2 diabetes mellitus of the consensus statement on carbohydrate-restricted diets by Korean Diabetes Association, Korean Society for the Study of Obesity, and Korean Society of Hypertension. Diabetes Metab J. 2022;46(3):377. doi:10.4093/DMJ.2022.0051

46. Jayedi A, Zeraattalab-Motlagh S, Jabbarzadeh B, et al. Dose-dependent effect of carbohydrate restriction for type 2 diabetes management: a systematic review and dose-response meta-analysis of randomized controlled trials. Am J Clin Nutr. 2022;116(1). doi:10.1093/AJCN/NQAC066

47. Strombotne KL, Lum J, Ndugga NJ, et al. Effectiveness of a ketogenic diet and virtual coaching intervention for patients with diabetes: a difference-in-differences analysis. Diabetes Obes Metab. 2021;23(12):2643-2650. doi:10.1111/DOM.14515

48. Virta Health. Virta Health highlights lasting, transformative health improvements in 5-year diabetes reversal study. June 5, 2022. Accessed October 6, 2023. https://www.virtahealth.com/blog/virta-sustainable-health-improvements-5-year-diabetes-reversal-study

49. Wan Z, Shan Z, Geng T, et al. Associations of moderate low-carbohydrate diets with mortality among patients with type 2 diabetes: a prospective cohort study. J Clin Endocrinol Metab. 2022;107(7):E2702-E2709. doi:10.1210/CLINEM/DGAC235

50. Akter S, Mizoue T, Nanri A, et al. Low carbohydrate diet and all cause and cause-specific mortality. Clin Nutr. 2021;40(4):2016-2024. doi:10.1016/J.CLNU.2020.09.022

51. Shan Z, Guo Y, Hu FB, Liu L, Qi Q. Association of low-carbohydrate and low-fat diets with mortality among US adults. JAMA Intern Med. 2020;180(4):513-523. doi:10.1001/JAMAINTERNMED.2019.6980

Issue
Federal Practitioner - 41(1)a
Issue
Federal Practitioner - 41(1)a
Page Number
6
Page Number
6
Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>0124 FED Keto</fileName> <TBEID>0C02EE34.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EE34</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240103T212824</firstPublished> <LastPublished>20240103T212824</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240103T212824</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Robert C. Oh, MD, MPHa; Kendrick C. Murphy, PharmD, BCACP, MHPb; Cory M. Jenks, PharmD, MHP, BCPS, BCACPc; Kathleen B. Lopez, RDN, CDCES, CNSCd; Mahendra A. Patel, PharmD, BCPSe; Emily E. Scotland, MSN, FNP-Ce; Monu Khanna, MD, MHPf</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>The prevalence of diabetes continues to increase despite advances in treatment options. In 2019, according to the Centers for Disease Control and Prevention (CD</metaDescription> <articlePDF/> <teaserImage/> <title>Low-Carbohydrate and Ketogenic Dietary Patterns for Type 2 Diabetes Management</title> <deck/> <eyebrow>Clinical Review</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>January</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>1</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2967</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>January 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Clinical Review | 2967<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">49</term> </sections> <topics> <term canonical="true">205</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Low-Carbohydrate and Ketogenic Dietary Patterns for Type 2 Diabetes Management</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Type 2 diabetes mellitus (T2DM) has been traditionally considered a chronic, progressive disease. Since 2017, guidelines from the US Department of Veterans Affairs and US Department of Defense have included low-carbohydrate (LC) dietary patterns in managing T2DM. Recently, carbohydrate reduction, including ketogenic diets, has gained renewed interest in the management and remission of T2DM. <br/><br/><b>Observations:</b> This narrative review examines the evidence behind carbohydrate reduction in T2DM and a practical guide for clinicians starting patients on therapeutic LC diets. We present an illustrative case and provide practical approaches to prescribing a very LC ketogenic (&lt; 50 g), LC (50-100 g), or a moderate LC (101-150 g) dietary plan and discuss adverse effects and management of LC diets. We provide a medication management and deprescription approach and discuss strategies to consider in conjunction with LC diets. As patients adopt LC diets, glycemia improves, and medications are deprescribed, hemoglobin A<sub>1c</sub> levels and fasting glucose may drop below the diagnostic threshold for T2DM. Remission of T2DM may occur with LC diets (hemoglobin A<sub>1c</sub> &lt; 6.5% for ≥ 3 months without T2DM medications). Finally, we describe barriers and limitations to applying therapeutic carbohydrate reduction in a federal health care system. <br/><br/><b>Conclusions:</b> The effective use of LC diets with close and intensive lifestyle counseling and a safe approach to medication management and deprescribing can improve glycemic control, reduce the overall need for insulin and medication and provide sustained weight loss. The efficacy and continuation of therapeutic carbohydrate reduction for patients with T2DM appears promising. Further research on LC diets, emerging strategies, and long-term effects on cardiometabolic risk factors, morbidity, and mortality will continue to inform practice.</p> <p><span class="Drop">T</span>he prevalence of diabetes continues to increase despite advances in treatment options. In 2019, according to the Centers for Disease Control and Prevention (CDC), 37.1 million (14.7%) US adults had diabetes. Among adults aged ≥ 65 years, the prevalence is even higher at 29.2%.<sup>1</sup> Research has also estimated that 45% of adults have evidence of prediabetes or diabetes.<sup>2</sup> According to the Veterans Health Administration, almost 25% of enrolled veterans have diabetes.<sup>3</sup></p> <h2>Background</h2> <p>Diabetes is associated with an increased risk of microvascular complications (eg, retinopathy, nephropathy, and neuropathy) and macrovascular complications (eg, atherosclerotic cardiovascular disease) and is one of the most common causes of morbidity and mortality in the US.<sup>4</sup> In 2017, diabetes was estimated to cost $327 billion in the US, up from $261 billion in 2012.<sup>5</sup> During this same period, the excess costs per person with diabetes increased from $8417 to $9601.<sup>5</sup></p> <p>Type 2 diabetes mellitus (T2DM) and its associated insulin resistance is typically considered a chronic disease with progressive loss of β-cell function. Controlling glycemia, delaying microvascular changes, and preventing macrovascular disease are major management goals. Lifestyle interventions are essential in the management and prevention of T2DM. Medication management for T2DM usually progresses through several medications, ending in insulin therapy.<sup>6</sup> Within 10 years of diagnosis, almost half of all individuals with T2DM will require insulin to manage their glycemia.<sup>7<br/><br/></sup>Bariatric surgery and nutrition approaches have been successful in reversing T2DM. Recently, there has been increased interest in nutritional approaches to place T2DM in remission, reverse the disease process, and improve insulin resistance. Contrary to popular belief, before the discovery of insulin in 1921, low-carbohydrate (LC) diets were the most common treatment for T2DM.<sup>8</sup> With the discovery of insulin and the eventual development of low-fat dietary recommendations, LC diets were no longer favored by most clinicians.<sup>8</sup> Low-fat diets are, by definition, also high-carbohydrate diets. By the early 1980s, low-fat diets had become the standard of care dietary recommendation, and the goal for clinicians became glycemic maintenance (with increased use of medications) rather than preventing hyperglycemia.<sup>8<br/><br/></sup>With growing evidence regarding the use of LC diets for T2DM, the US Department of Veterans Affairs (VA) and US Department of Defense (DoD), the American Diabetes Association (ADA), the European Association for the Study of Diabetes (EASD), Diabetes Canada, and Diabetes Australia all include LC diets as a viable option for treating T2DM.<sup>4,9-12</sup> This article will highlight a case using a reduced carbohydrate approach in lifestyle management and provide clinicians with practical guidance in its implementation. We will review the evidence that informs these guidelines, describe a practical approach to nutritional counseling, and review medication management and deprescribing approaches. Finally, barriers to implementation will be explored.</p> <h2>ILLUSTRATIVE CASE</h2> <p>A 64-year-old woman presented to the clinical pharmacist for the management of T2DM after her tenth hospitalization related to hyperglycemia in 10 years. She had previously been managed by primary care clinicians, clinical dietitians, endocrinologists, and certified diabetes care and education specialists. Pertinent history included diabetic ketoacidosis, coronary artery disease, hyperlipidemia, hypertension, obstructive sleep apnea, obesity, metabolic dysfunction-associated steatotic liver disease, and mild nonproliferative diabetic retinopathy with clinically significant macular edema. The patient expressed frustration with poor glycemic control during her many years of insulin therapy and an inability to lose weight due to insulin dose titrations. The patient reported prior education including but not limited to standardized sample menus, consistent carbohydrate intake, calorie reduction, general healthful nutrition, and the “move more, eat less” approach. The patient was unable to titrate insulin dosage and did not experience weight loss despite compliance with these methods.</p> <p>Her medications included glargine insulin 45 units once daily, aspart insulin 5 units before meals 3 times daily, and metformin 1000 mg twice daily. Her hemoglobin A<sub>1c</sub> (HbA<sub>1c</sub>) level was 11.8%. A review of prior therapies for T2DM included glyburide 5 mg twice daily, metformin 1000 mg twice daily, 70/30 insulin (up to 340 units/d), glargine insulin (range, 10-140 units/d), regular insulin (range, 30-240 units/d), aspart insulin (range, 15-45 units/d), and U-500 regular insulin (range, 125-390 units/d). She took metoprolol 25 mg extended release daily and hydrochlorothiazide 25 mg daily, but both were discontinued after the most recent hospitalization. A review of HbA<sub>1c</sub> readings showed poor glycemic control for &gt; 12 years (range, 10.3% to &gt; 12.3%).Education for lifestyle modifications, including an LC diet, was presented to the patient to assist with weight loss, improve glycemic control, and reduce insulin resistance. In addition, a glucagon-like peptide-1 agonist (liraglutide) was added to her pharmacotherapy. Continued dietary modifications with LC intake led to consistent reductions in glargine and aspart insulin therapy. The patient remained motivated throughout clinic visits due to improved glycemic control with sustainable dietary modifications, consistently reported feeling better overall, and deprescribed diabetes drug therapies. She remained off her blood pressure medications. After4 months of LC dietary modifications, all insulin therapy was discontinued. She continued with liraglutide 1.8 mg daily and metformin 1000 mg twice daily with an HbA<sub>1c</sub> of 6.3%. Two months later, her HbA<sub>1c</sub> level was 6.0%. She also lost 8 lb and her body mass index improved from 31 to 29.</p> <h2>Low-Carbohydrate T2DM DIET MANAGEMENT</h2> <p>LC diets are commonly defined as &lt; 130 g of carbohydrates per day.<sup>13</sup> Very LC ketogenic (VLCK) diets often contain ≤ 50 g of carbohydrates per day to induce nutritional ketosis.<sup>13</sup> One of the first randomized controlled trials (RCTs) that compared a VLCK diet (&lt; 30 g of carbohydrates per day) with a low-fat diet for obesity demonstrated greater weight loss at 6 months with the LC diet. In addition, patients with diabetes randomized to the LC group also showed improved insulin sensitivity. Notably, this study was done in a population of veterans enrolled at the VA Philadelphia Health Care System.<sup>14</sup> </p> <p>A 2008 study comparing an LC diet with a calorie-restricted, low-glycemic diet for individuals with T2DM found that the LC diet group experienced a greater reduction in HbA<sub>1c</sub> and insulin levels and weight.<sup>15</sup> Comparing these 2 diet groups after 24 weeks, 95% of individuals in the LC group reduced or discontinued T2DM medications vs 62% in the low-glycemic group.<sup>15</sup> Another study of individuals with T2DM compared a VLCK diet with a low-fat diet. After 34 weeks, 55% of individuals in the LC diet group achieved an HbA<sub>1c</sub> level below the threshold for diabetes vs 0% in the low-fat diet group.<sup>16</sup> A 2018 study of patients with T2DM investigated the impact of a very LC diet compared with the standard of care.<sup>17</sup> After 1 year, the LC diet group experienced a mean HbA<sub>1c</sub> reduction of 1.3%, and 60% of individuals who completed the study achieved an HbA<sub>1c</sub> level &lt; 6.5% without T2DM medications (not including metformin). This study also demonstrated that medications were significantly reduced, including 100% discontinuation of sulfonylureas and 94% reduction or elimination of insulin.<br/><br/>A recent study of an LC diet (&lt; 20% energy from carbohydrates) demonstrated reduced HbA<sub>1c</sub> levels, weight, and waist circumference vs a control diet after 6 months. The control diet derived 50% to 60% of energy from carbohydrates.<sup>18</sup> This study is typical of other LC interventions, which did not calorie restrict and instead allowed ad libitum intake.<sup>14,15<br/><br/></sup>With mounting evidence, the VA/DoD guidelines on T2DM management included LC diets as dietary options for treating T2DM. The ADA also determined that LC diets had the most evidence in improving glycemia and included LC diets as an option for medical nutrition therapy (Table 1).<sup>10,19<br/><br/></sup>A systematic review and meta-analysis looking at RCTs of LC diets found evidence for remission of T2DM without significant adverse effects (AEs).<sup>20</sup> Another recent systematic review and network meta-analysis of 42 RCTs found that the ketogenic diet was superior for a reduction in HbA<sub>1c</sub> levels compared with 9 other dietary patterns, including low-fat, Mediterranean, and vegetarian/vegan diets. Overall, ketogenic, Mediterranean, moderate-carbohydrate, and low-glycemic index diets demonstrated improved glycemic control.<sup>21<br/><br/></sup>Ideally, a comprehensive behavioral program, such as the VA Move! or Whole Health program, should incorporate patient aligned care teams (PACTs), behavioral health clinicians, clinical pharmacists, and dietitians to provide medical-nutrition therapy using LC diets. However, many facilities may not have adequate experience, expertise, or support. We provide practical approaches to provide LC nutrition counseling, medication management, and deprescribing for any primary care clinician applying LC diets for their patients. For simplicity and practicality, we define 3 types of LC dietary patterns: (1) VLCK (&lt; 50 g); (2) LC (50-100 g); and (3) moderate LC (101-150 g).</p> <h2>Nutrition</h2> <p>All nutrition approaches, including LC diets, should be patient centered, individualized, and sensitive to the patient's culture. Typically, many patients have previously been instructed to consume low-fat (and subsequently) high-carbohydrate (&gt; 150 g) meals. Most well-meaning clinicians have provided common-approach diet education from mainstream health organizations in the form of standardized handouts. For example, the Carbohydrate Counting for People with Diabetes patient education handout from the Academy of Nutrition and Dietetics provides a sample menu with 3 meals and 1 snack totaling 195 g of carbohydrates.<sup>22</sup> In contrast, an example ADA diet has sample diets with 3 meals and 2 snacks with approximately 20 to 70 g of carbohydrates.<sup>23</sup> In the VA, there are excellent resources to review and standardize handouts that emphasize an LC nutrition approach to T2DM, including ketogenic versions.<sup>24,25</sup> Table 2 shows example meal plans based on different LC patterns—VLCK, LC, and moderate LC.</p> <p>Starting an LC dietary pattern should maximize nutrient-dense and minimally processed proteins. Clinicians should begin with a baseline nutritional assessment through a 24-hour recall or food diary. After this has been completed, the patient’s baseline diet is assessed, and a gradual carbohydrate reduction plan is discussed. Generally, carbohydrate reduction is recommended at 1 meal per day per week. High-carbohydrate meals and snacks are restructured to favor satiating, minimally processed, high-protein food sources. Individual food preferences are considered and included in the recommended LC plan. For example, LC diets can be formulated for vegetarians and vegans as well as those who prefer meat and seafood. Prioritizing satiating and nutrient-dense foods can help increase the probability of diet acceptance and adherence. <br/><br/>A recent study<sup> </sup>showed that restricting carbohydrates at breakfast reduces 24-hour postprandial hyperglycemia and improves glycemic variability.<sup>26</sup> Many patients consume upward of 50 g of carbohydrates at breakfast.<sup>27</sup> For example, it is not uncommon for a patient to consume cereal with milk or oatmeal, orange juice, a banana, and toast at breakfast. Instead, the patient is advised to consume any combination of eggs, meat, no-sugar-added Greek yogurt, or berries. <br/><br/>To keep things simple for lunch and dinner, the patient is offered high-quality, minimally processed protein of their choosing with any nonstarchy vegetable. Should a patient desire additional carbohydrates with meals, they may reduce the baseline serving of carbohydrates by 50%. For example, if a patient normally fills 50% of their plate with spaghetti, they may reduce the pasta portion to 25% and add a meatball or increase the amount of vegetables consumed with the meal to satiety.<br/><br/>Snacks may include cheese, eggs, peanut butter, nuts, seeds, berries, no-sugar-added Greek yogurt, or guacamole. Oftentimes, when LC meals are adopted, the desire or need for snacking is diminished due to the satiating effect of high-quality protein sources and nonstarchy vegetables.</p> <h3>Adverse Effects</h3> <p>AEs have been reported with VLCK diets, including headache, diarrhea, constipation, muscle cramps, halitosis, light-headedness, and muscle weakness.<sup>28</sup> These AEs may be mitigated with increased fluid intake, sodium intake, and magnesium supplementation.<sup>29</sup> Increasing fluids to a minimum of 2 L/d and adding sodium (eg, bouillon supplementation) can minimize AEs.<sup>30</sup> Milk of magnesia (5 mL) or slow-release magnesium chloride 200 mEq/d is suggested to reduce muscle cramps.<sup>30</sup> There have been no studies looking at sodium intake and worsening hypertension or chronic heart failure in the setting of an LC diet, but fluid and electrolyte intake should be monitored closely, especially in patients with uncontrolled hypertension and heart failure. Other concerns of higher protein on worsening kidney function have generally not been founded.<sup>31</sup> In some individuals, an LC and higher fat diet may increase low-density lipoprotein cholesterol (LDL-C).<sup>32</sup> Therefore a baseline lipid panel is recommended and should be monitored along with HbA<sub>1c</sub> levels. An elevated LDL-C response may be managed by increasing protein and reducing saturated fat intake while maintaining the reduced carbohydrate content of the diet. </p> <h2>Medication Management</h2> <p>The adoption of an LC diet can cause a swift and profound reduction in blood sugar.<sup>33</sup> Utilizing PACTs can help prevent adverse drug events by involving clinical pharmacists to provide recommendations and dose reductions as patients adopt an LC diet. Each approach must be individualized to the patient and can depend on several factors, including the number and strength of medications, the degree of carbohydrate reduction, baseline blood glucose, as well as assessing for medical literacy and ability to implement recommendations. Additionally, patients should monitor their blood sugar regularly and communicate with their primary care team (pharmacist, PACT registered nurse, primary care clinician, and registered dietician). Ultimately, the goal when adopting an LC diet while taking antihyperglycemics is safely avoiding hypoglycemia while reducing the number of medications the patient is taking. We summarize a practical approach to medication management that was recently published (Table 3).<sup>33,34</sup> </p> <h3>Medications to Reduce or Discontinue</h3> <p>Medications that can cause hypoglycemia should be the first to be reduced or discontinued upon starting an LC diet, including bolus insulin (although a small amount may be needed to correct for high blood sugar), sulfonylureas, and meglitinides. Combination insulin should be stopped and changed to basal insulin to avoid the risk of hypoglycemia (see Table 4 for insulin deprescribing recommendations). The mechanism of action in preventing the breakdown of carbohydrates in the gastrointestinal tract makes the use of α-glucosidase inhibitors superfluous, and they can be discontinued, reducing pill burden and polypharmacy risks. Sodium-glucose transport protein 2 inhibitors (SGLT2i) should be discontinued for patients on VLCK diets due to the risk of euglycemic diabetic ketoacidosis. However, with LC and moderate LC plans, the SGLT2i may be used with caution as long as patients are made aware of ketoacidosis symptoms. To help prevent the risk of hypoglycemia, basal/long-acting insulin can be continued, but at a 50% reduced dose. Patients should closely monitor blood sugar to assess for appropriateness of dose reductions. While thiazolidinediones are not contraindicated, clinicians can consider discontinuation given both their penchant for inducing weight gain and their limited outcomes data.</p> <h3>Medications to Continue</h3> <p>Medications that pose minimal risk for hypoglycemia can be continued, including metformin, dipeptidyl peptidase 4 inhibitors, and glucagon-like peptide-1 agonists. However, even though these may pose a low risk of hypoglycemia, patients should still closely monitor their blood glucose so medications can be deprescribed as soon as safely and reasonably possible.</p> <h3>Other Medications</h3> <p>The improvement in metabolic health with the reduction of carbohydrates can render other classes of medications unnecessary or require adjustment. Patients should be counseled to monitor their blood pressure as significant and rapid improvements can occur. In the event of a systolic blood pressure of 100 to 110 mm Hg or signs of hypotension, down titration or discontinuation of antihypertensives should be initiated. Limited evidence exists on the preferred order of discontinuation but should be informed by other comorbidities, such as coronary artery disease and chronic kidney disease. Given an LC diet’s diuretic effect, tapering and stopping diuretics may be an option. Other medications requiring closer monitoring include lithium (can be affected by fluid and electrolyte shifts), warfarin (may alter vitamin K intake), valproate (which may be reduced), and zonisamide and topiramate (kidney stone risk).</p> <h2>Remission of T2DM with LC Diets</h2> <p>As patients adopt LC diets and medications are deprescribed and glycemia improves, HbA<sub>1c</sub> and fasting glucose levels may drop below the diagnostic threshold for T2DM.<sup>20</sup> As new evidence emerges surrounding the management of T2DM from a lifestyle perspective, major health care organizations have acknowledged that T2DM is not necessarily an incurable, progressive disease, but rather a disease that can be reversed or put in remission.<sup>35-37</sup> In 2016, the World Health Organization (WHO) global report on diabetes acknowledged that T2DM reversal can be achieved via weight loss and calorie restriction.<sup>35</sup></p> <p>In 2021, a consensus statement from the ADA, the Endocrine Society, the EASD, and Diabetes UK defined T2DM remission as an HbA<sub>1c</sub> level &lt; 6.5% for at least 3 months with no T2DM medications.<sup>36</sup> Diabetes Australia also published a position statement in 2021 about T2DM remission.<sup>37</sup> Like the WHO, Diabetes Australia acknowledged that remission of T2DM is possible following intensive dietary changes or bariatric surgery.<sup>37</sup> Before the 2021 consensus statement, some experts argued that excluding metformin from the T2DM medication list may not be warranted since metformin has indications beyond T2DM. In this case, remission of T2DM could be defined as an HbA<sub>1c</sub> level &lt; 6.5% for at least 3 months and on metformin or no T2DM medications.<sup>8</sup>  </p> <h2>Emerging Strategies</h2> <p>Emerging strategies, such as continuous glucose monitors (CGMs) and the use of intermittent fasting/time-restricted eating (TRE), can be used with the LC diet to help improve the monitoring and management of T2DM. In the recently published VA/DoD guidelines for T2DM, the work group suggested real-time CGMs for qualified patients with T2DM.<sup>4</sup> These include patients on daily insulin who are not achieving glycemic control or to reduce the risk for hypoglycemia. CGMs have shown evidence of improved glycemic control and decreased hypoglycemia in those with T2DM.<sup>38,39</sup> It is currently unknown if CGMs improve long-term glycemic control, but they appear promising for managing and reducing medications for those on an LC diet.<sup>40</sup></p> <p>TRE can be supplemented with an LC plan that incorporates “eating windows.” Common patterns include 14 hours of fasting and a 10-hour eating window (14F:10E), or 16 hours of fasting and an 8-hour eating window (16F:8E). By eating only in the specified window, patients generally reduce caloric intake and minimize insulin and glucose excursions during the fasting window. No changes need to be made to the macronutrient composition of the diet, and LC approaches can be used with TRE. The mechanism of action is likely multifactorial, targeting hyperinsulinemia and insulin resistance as well as producing a caloric deficit to enable weight loss.<sup>41</sup> Eating windows may improve insulin sensitivity, reduce insulin resistance, and enhance overall glycemic control. The recent VA/DoD guidelines recommended against intermittent fasting due to concerns over the risk of hypoglycemia despite larger weight loss in TRE groups.<sup>4</sup> Recently, a study using CGMs and TRE demonstrated both improved glycemic control and no hypoglycemic episodes in patients with T2DM on insulin.<sup>42</sup> Patients who would like to supplement TRE with an LC plan as a strategy for improved glycemic control should work closely with their PACT to help manage their TRE and LC plan and consider a CGM adjunct, especially if on insulin.</p> <h2>Barriers</h2> <p>Managing T2DM often requires comprehensive lifestyle modifications of nutrition, exercise, sleep, stress management, and other psychosocial issues, as well as an interdisciplinary team-based approach.<sup>43</sup> The advantage of working within the VA includes a uniform system within a network of care. However, many patients continue to use both federal and private health care. This use of out-of-network care may result in fragmented, potentially disjointed, or even contradictory dietary advice.</p> <p>The VA PACT, whole health for holistic health, and weight loss interventions such as the MOVE! program provide lifestyle interventions like nutrition, physical activity, and behavior change. However, these well-intentioned approaches may provide alternative and even diverging recommendations, which place additional barriers to effective patient management. In patients who are advised and accept a trial of an LC plan, each member of the team should embrace the self-management decision of the patient and support the plan.<sup>29</sup> Any conflicts, questions, or concerns should be communicated directly with the team in an interdisciplinary approach to provide a unified message and counsel.<br/><br/>The long-term effects and sustainability of an LC diet have been questioned in the literature.<sup>44-46</sup> Recently, the use of an app-based coaching plan has demonstrated short- and long-term sustainability on an LC diet.<sup>47</sup> In just 5 months in a large VA system, 590 patients using a virtual coaching platform and a VLCK diet plan were found to have lower HbA<sub>1c</sub> levels, reduced diabetic medication fills, lower body mass index, fewer outpatient visits, and lower prescription drug costs. <br/><br/>A 5-year follow-up found nearly 50% of participants sustained a VLCK diet for T2DM. For patients who participated in the study after 2 years, 72% sustained the VLCK diet in years 2 to 5. Most required nearly 50% fewer medications and in those that started with insulin, half did not require it at 5 years.<sup>48</sup> Further research, however, is necessary to determine the long-term effects on cardiometabolic markers and health with LC diets. There are no long-term RCTs on outcomes data looking at T2DM morbidity or mortality. While there are prospective cohort studies on LC diets in the general population on mortality, they demonstrate mixed results. These studies may be confounded by heterogeneous definitions of LC diets, diet quality, and other health factors.<sup>49-51</sup></p> <h2>Conclusions</h2> <p>The effective use of LC diets within a PACT with close and intensive lifestyle counseling and a safe approach to medication management and deprescribing can improve glycemic control, reduce the overall need for insulin, reduce medication use, and provide sustained weight loss. Additionally, the use of therapeutic carbohydrate reduction and subsequent medication deprescription may lead to sustained remission of T2DM. The current efficacy and sustainment of therapeutic carbohydrate reduction for patients with T2DM appears promising. Further research on LC diets, emerging strategies, and long-term effects on cardiometabolic risk factors, morbidity, and mortality will continue to inform future practice in our health care system.</p> <h3> Acknowledgments </h3> <p> <em>We thank Cecile Seth who has been instrumental in pushing us forward and the Metabolic Multiplier group who has helped encourage and provide input into this article.</em> </p> <h3> Author affiliations </h3> <p> <em><sup>a</sup>Veterans Affairs Palo Alto Health Care System, California<br/><br/><sup>b</sup>Western North Carolina Veterans Affairs Health Care System, Asheville<br/><br/><sup>c</sup>Ambulatory Care Clinical Pharmacist Society of Metabolic Health Practitioners, Tucson, Arizona<br/><br/><sup>d</sup>Veterans Affairs Boston Health Care System, Massachusetts<br/><br/><sup>e</sup>Southern Arizona Veterans Affairs Health Care System, Tucson<br/><br/><sup>f</sup>Veterans Affairs St Louis Health Care System, Missouri</em> </p> <h3> Author disclosures </h3> <p> <em>CM Jenks is married to an employee of Virta Medical, which provides care related to type 2 diabetes and ketogenic diets. </em> </p> <h3> Disclaimer </h3> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner</i>, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.</em> </p> <h3> Ethics and consent </h3> <p> <em>Written consent for publication has been obtained from the patient reported in the illustrative case.</em> </p> <h3> References </h3> <p class="reference"> 1. Centers for Disease Control and Prevention. Prevalence of Both Diagnosed and Undiagnosed Diabetes. Updated September 30, 2022. Accessed October 6, 2023. https://www.cdc.gov/diabetes/data/statistics-report/diagnosed-undiagnosed-diabetes.html<br/><br/> 2. Centers for Disease Control and Prevention. Diabetes and Prediabetes. Updated September 6, 2022. Accessed October 6, 2023. https://www.cdc.gov/chronicdisease/resources/publications/factsheets/diabetes-prediabetes.htm 3. US Department of Veterans Affairs. Diabetes information - Nutrition and food services. Updated May 4, 2023. Accessed October 6, 2023. https://www.nutrition.va.gov/diabetes.asp<br/><br/> 4. US Department of Veterans Affairs. Management of Type 2 Diabetes Mellitus (2023) - VA/DoD Clinical Practice Guidelines. Updated September 1, 2023. Accessed October 6, 2023. https://www.healthquality.va.gov/guidelines/CD/diabetes/<br/><br/> 5. American Diabetes Association. Economic Costs of Diabetes in the U.S. in 2017. <i>Diabetes Care</i>. 2018;41(5):917-928. doi:10.2337/dci18-0007<br/><br/> 6. Home P, Riddle M, Cefalu WT, et al. Insulin therapy in people with type 2 diabetes: opportunities and challenges?. <i>Diabetes Care</i>. 2014;37(6):1499-1508. doi:10.2337/dc13-2743<br/><br/> 7. Donath MY, Ehses JA, Maedler K, et al. Mechanisms of β-cell death in type 2 diabetes. <i>Diabetes</i>. 2005;54(suppl 2):S108-S113. doi:10.2337/DIABETES.54.SUPPL_2.S108<br/><br/> 8. Hallberg SJ, Gershuni VM, Hazbun TL, Athinarayanan SJ. Reversing type 2 diabetes: a narrative review of the evidence. <i>Nutrients</i>. 2019;11(4):766. Published 2019 Apr 1. doi:10.3390/nu11040766<br/><br/> 9. Davies MJ, D’Alessio DA, Fradkin J, et al. Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). <i>Diabetes Care</i>. 2018;41(12):2669. doi:10.2337/DCI18-0033<br/><br/>10. Evert AB, Dennison M, Gardner CD, et al. Nutrition therapy for adults with diabetes or prediabetes: a consensus report. <i>Diabetes Care</i>. 2019;42(5):731-754. doi:10.2337/DCI19-0014<br/><br/>11. Diabetes Canada position statement on low-carbohydrate diets for adults with diabetes: a rapid review. <i>Can J Diabetes</i>. 2020;44(4):295-299. doi:10.1016/J.JCJD.2020.04.001<br/><br/>12. Diabetes Australia. Position statements. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/research-advocacy/position-statements/<br/><br/>13. Feinman RD, Pogozelski WK, Astrup A, et al. Dietary carbohydrate restriction as the first approach in diabetes management: critical review and evidence base. <i>Nutrition</i>. 2014;31(1):1-13. doi:10.1016/j.nut.2014.06.011<br/><br/>14. Samaha FF, Iqbal N, Seshadri P, et al. A low-carbohydrate as compared with a low-fat diet in severe obesity. <i>N Engl J Med</i>. 2003;348(21):2074-2081. doi:10.1056/NEJMOA02263715. Westman EC, Yancy WS, Mavropoulos JC, Marquart M, McDuffie JR. The effect of a low-carbohydrate, ketogenic diet versus a low-glycemic index diet on glycemic control in type 2 diabetes mellitus. <i>Nutr Metab (Lond)</i>. 2008;5(1):36. doi:10.1186/1743-7075-5-36<br/><br/>16. Saslow LR, Mason AE, Kim S, et al. An online intervention comparing a very low-carbohydrate ketogenic diet and lifestyle recommendations versus a plate method diet in overweight individuals with type 2 diabetes: a randomized controlled trial. <i>J Med Internet Res</i>. 2017;19(2). doi:10.2196/JMIR.5806<br/><br/>17. Hallberg SJ, McKenzie AL, Williams PT, et al. Effectiveness and safety of a novel care model for the management of type 2 diabetes at 1 year: an open-label, non-randomized, controlled study. <i>Diabetes Ther</i>. 2018;9(2):583-612. doi:10.1007/S13300-018-0373-9<br/><br/>18. Gram-Kampmann EM, Hansen CD, Hugger MB, et al. Effects of a 6-month, low-carbohydrate diet on glycaemic control, body composition, and cardiovascular risk factors in patients with type 2 diabetes: An open-label randomized controlled trial. <i>Diabetes Obes Metab</i>. 2022;24(4):693-703. doi:10.1111/DOM.14633<br/><br/>19. Committee ADAPP. 5. Facilitating behavior change and well-being to improve health outcomes: standards of medical care in diabetes—2022. <i>Diabetes Care</i>. 2022;45(suppl 1):S60-S82. doi:10.2337/DC22-S005<br/><br/>20. Goldenberg JZ, Johnston BC. Low and very low carbohydrate diets for diabetes remission. <i>BMJ</i>. 2021;373:m4743. doi:10.1136/BMJ.N262</p> <p class="reference">21. Jing T, Zhang S, Bai M, et al. Effect of dietary approaches on glycemic control in patients with type 2 diabetes: a systematic review with network meta-analysis of randomized trials. <i>Nutrients</i>. 2023;15(14):3156. doi:10.3390/nu15143156<br/><br/>22. Academy of Nutrition and Dietetics. Nutrition care manual. Accessed October 6, 2023. https://www.nutritioncaremanual.org/<br/><br/>23. Low carbohydrate and very low carbohydrate eating patterns in adults with diabetes. ShopDiabetes.org. Accessed August 5, 2022. https://shopdiabetes.org/products/low-carbohydrate-and-very-low-carbohydrate-eating-patterns-in-adults-with-diabetes-a-guide-for-health-care-providers<br/><br/>24. US Department of Veterans Affairs. Diabetes education - nutrition and food services. Published July 31, 2022. http://vaww.nutrition.va.gov/docs/pted/ModifiedKetogenicDiet.pdf [Source not verified] <br/><br/>25. US Department of Veterans Affairs, My HealtheVet. Lowdown on low-carb diets. Updated June 1, 2021. Accessed October 6, 2023. https://www.myhealth.va.gov/mhv-portal-web/ss20190724-low-carb-diet<br/><br/>26. Chang CR, Francois ME, Little JP. Restricting carbohydrates at breakfast is sufficient to reduce 24-hour exposure to postprandial hyperglycemia and improve glycemic variability. <i>Am J Clin Nutr</i>. 2019;109(5):1302-1309. doi:10.1093/AJCN/NQY261<br/><br/>27. Hall KD, Ayuketah A, Brychta R, et al. Ultra-processed diets cause excess calorie intake and weight gain: an inpatient randomized controlled trial of ad libitum food intake. <i>Cell Metab</i>. 2019;30(1):226. doi:10.1016/j.cmet.2019.05.020<br/><br/>28. Harvey CJ d. C, Schofield GM, Zinn C, Thornley S. Effects of differing levels of carbohydrate restriction on mood achievement of nutritional ketosis, and symptoms of carbohydrate withdrawal in healthy adults: a randomized clinical trial. <i>Nutrition</i>. 2019;67-68:100005. doi:10.1016/J.NUTX.2019.100005<br/><br/>29. Griauzde DH, Standafer Lopez K, Saslow LR, Richardson CR. A pragmatic approach to translating low- and very low-carbohydrate diets into clinical practice for patients with obesity and type 2 diabetes. <i>Front Nutr</i>. 2021;8:416. doi:10.3389/FNUT.2021.682137/BIBTEX<br/><br/>30. Westman EC, Tondt J, Maguire E, Yancy WS. Implementing a low-carbohydrate, ketogenic diet to manage type 2 diabetes mellitus. <i>Expert Rev Endocrinol Metab</i>. 2018;13(5):263-272. doi:10.1080/17446651.2018.1523713<br/><br/>31. Suyoto PST. Effect of low-carbohydrate diet on markers of renal function in patients with type 2 diabetes: a meta-analysis. <i>Diabetes Metab Res Rev</i>. 2018;34(7). doi:10.1002/DMRR.3032<br/><br/>32. Norwitz NG, Feldman D, Soto-Mota A, Kalayjian T, Ludwig DS. Elevated LDL cholesterol with a carbohydrate-restricted diet: evidence for a “lean mass hyper-responder” phenotype. <i>Curr Dev Nutr</i>. 2021;6(1). doi:10.1093/CDN/NZAB144<br/><br/>33. Murdoch C, Unwin D, Cavan D, Cucuzzella M, Patel M. Adapting diabetes medication for low carbohydrate management of type 2 diabetes: a practical guide. <i>Br J Gen Pract</i>. 2019;69(684):360-361. doi:10.3399/bjgp19X704525<br/><br/>34. Cucuzzella M, Riley K, Isaacs D. Adapting medication for type 2 diabetes to a low carbohydrate diet. <i>Front Nutr</i>. 2021;8:486. doi:10.3389/FNUT.2021.688540/BIBTEX<br/><br/>35. World Health Organization. Global report on diabetes. 2016. Accessed October 6, 2023. https://iris.who.int/bitstream/handle/10665/204871/9789241565257_eng.pdf?sequence=1<br/><br/>36. Riddle MC, Cefalu WT, Evans PH, et al. Consensus report: definition and interpretation of remission in type 2 diabetes. <i>Diabetes Care</i>. 2021;44(10):2438-2444. doi:10.2337/DCI21-0034</p> <p class="reference">37. Diabetes Australia. Type 2 Diabetes remission position statement. 2021. Accessed October 6, 2023. https://www.diabetesaustralia.com.au/wp-content/uploads/2021_Diabetes-Australia-Position-Statement_Type-2-diabetes-remission_2.pdf<br/><br/>38. Martens T, Beck RW, Bailey R, et al. Effect of continuous glucose monitoring on glycemic control in patients with type 2 diabetes treated with basal insulin: a randomized clinical trial. <i>JAMA</i>. 2021;325(22):2262-2272. doi:10.1001/JAMA.2021.7444<br/><br/>39. Jackson MA, Ahmann A, Shah VN. Type 2 diabetes and the use of real-time continuous glucose monitoring. <i>Diabetes Technol Ther</i>. 2021;23(S1):S27-S34. doi:10.1089/DIA.2021.0007<br/><br/>40. Oser TK, Cucuzzella M, Stasinopoulos M, Moncrief M, McCall A, Cox DJ. An innovative, paradigm-shifting lifestyle intervention to reduce glucose excursions with the use of continuous glucose monitoring to educate, motivate, and activate adults with newly diagnosed type 2 diabetes: pilot feasibility study. <i>JMIR Diabetes</i>. 2022;7(1). doi:10.2196/34465<br/><br/>41. Światkiewicz I, Woźniak A, Taub PR. Time-restricted eating and metabolic syndrome: current status and future perspectives. <i>Nutrients</i>. 2021;13(1):221. doi:10.3390/NU13010221<br/><br/>42. Obermayer A, Tripolt NJ, Pferschy PN, et al. Efficacy and safety of intermittent fasting in people with insulin-treated type 2 diabetes (INTERFAST-2)—a randomized controlled trial. <i>Diabetes Care</i>. 2023;46(2):463-468. doi:10.2337/dc22-1622<br/><br/>43. American Diabetes Association. 5. Lifestyle management: standards of medical care in diabetes—2019. <i>Diabetes Care</i>. 2019;42(suppl 1):S46-S60. doi:10.2337/DC19-S005<br/><br/>44. Li S, Ding L, Xiao X. Comparing the efficacy and safety of low-carbohydrate diets with low-fat diets for type 2 diabetes mellitus patients: a systematic review and meta-analysis of randomized clinical trials. <i>Int J Endocrinol</i>. 2021;2021:8521756. Published 2021 Dec 6. doi:10.1155/2021/8521756<br/><br/>45. Choi JH, Kang JH, Chon S. Comprehensive understanding for application in Korean patients with type 2 diabetes mellitus of the consensus statement on carbohydrate-restricted diets by Korean Diabetes Association, Korean Society for the Study of Obesity, and Korean Society of Hypertension. <i>Diabetes Metab J</i>. 2022;46(3):377. doi:10.4093/DMJ.2022.0051<br/><br/>46. Jayedi A, Zeraattalab-Motlagh S, Jabbarzadeh B, et al. Dose-dependent effect of carbohydrate restriction for type 2 diabetes management: a systematic review and dose-response meta-analysis of randomized controlled trials. <i>Am J Clin Nutr</i>. 2022;116(1). doi:10.1093/AJCN/NQAC066<br/><br/>47. Strombotne KL, Lum J, Ndugga NJ, et al. Effectiveness of a ketogenic diet and virtual coaching intervention for patients with diabetes: a difference-in-differences analysis. <i>Diabetes Obes Metab</i>. 2021;23(12):2643-2650. doi:10.1111/DOM.14515<br/><br/>48. Virta Health. Virta Health highlights lasting, transformative health improvements in 5-year diabetes reversal study. June 5, 2022. Accessed October 6, 2023. https://www.virtahealth.com/blog/virta-sustainable-health-improvements-5-year-diabetes-reversal-study<br/><br/>49. Wan Z, Shan Z, Geng T, et al. Associations of moderate low-carbohydrate diets with mortality among patients with type 2 diabetes: a prospective cohort study. <i>J Clin Endocrinol Metab</i>. 2022;107(7):E2702-E2709. doi:10.1210/CLINEM/DGAC235<br/><br/>50. Akter S, Mizoue T, Nanri A, et al. Low carbohydrate diet and all cause and cause-specific mortality. <i>Clin Nutr</i>. 2021;40(4):2016-2024. doi:10.1016/J.CLNU.2020.09.022<br/><br/>51. Shan Z, Guo Y, Hu FB, Liu L, Qi Q. Association of low-carbohydrate and low-fat diets with mortality among US adults. <i>JAMA Intern Med</i>. 2020;180(4):513-523. doi:10.1001/JAMAINTERNMED.2019.6980</p> </itemContent> </newsItem> </itemSet></root>
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media