Three Anomalies and a Complication: Ruptured Noncoronary Sinus of Valsalva Aneurysm, Atrial Septal Aneurysm, and Patent Foramen Ovale

Article Type
Changed
Thu, 04/26/2018 - 09:02
The confluence of atrial septal aneurysm and patent foramen ovale in noncoronary sinus of Valsalva has not been previously documented in the literature.

A 53 year-old white male with a past medical history of hypertension, hyperlipidemia, and former tobacco use was referred to the Dayton VAMC in Ohio for symptoms that included shortness of breath and a recent abnormal stress test. The patient reported no history of known coronary artery disease (CAD), congestive heart failure, or other cardiovascular diseases. The patient also reported no recent fever, bacterial blood infection, syphilis infection, recreational drug use, or chest trauma.

A physical examination was remarkable for grade 3/6 continuous murmur at the 5th interspace to the left of the sternum and a loud “pistol shot” sound heard over the femoral artery. The patient had jugular venous distension and 2+ leg edema bilaterally. His vital signs were normal, and laboratory blood tests showed normal hemoglobin level and kidney function.

An electrocardiogram showed nonspecific ST segment changes and a transthoracic echocardiogram (TTE) revealed a high-velocity jet in the right atrium (RA) above the tricuspid valve concerning for sinus of Valsalva aneurysm (SVA).

fed03406033_f1.png
A transesophageal echocardiogram (TEE) showed a “windsock” appearance of the noncoronary SVA with possible rupture into the RA (Figure 1) and atrial septal aneurysm (ASA) with more than 2-cm displacement beyond the plane of the atrial septum and a 2-mm patent foramen ovale (PFO) (Figure 2).
fed03406033_f2.png
fed03406033_f3.png

 

Right heart catheterization revealed elevated RA pressures with positive shunt study showing oxygen saturation step-up in the RA (Figure 3). Left heart hemodynamic measurement from an aortic approach to the distal part of the noncoronary cusp SVA revealed an RA pressure-tracing pattern consistent with rupture of the noncoronary SVA into the RA (Figure 4).

fed03406033_f4.png
Coronary angiography revealed single vessel CAD involving the proximal right coronary artery.

The primary diagnosis was of acute heart failure secondary to ruptured aneurysm of the noncoronary SVA into RA. The patient also received a secondary diagnosis of atrial septal aneurysm and PFO.

[embed:render:related:node:128884]

Treatment & Outcome

The patient was treated with aggressive diuresis and responded well to therapy. Considering the high mortality rate associated with a ruptured SVA, the patient was referred to a tertiary care center for surgical evaluation. He underwent repair of aorto-right atrial communication with a Cormatrix patch (Roswell, GA) from the aortic side and with primary closure from the right atrial side with resection of the windsock tract; coronary artery bypass graft x1 with right internal mammary artery to the right coronary artery; closure of the PFO with the Cormatrix patch.

The postoperative TEE confirmed preserved LV and RV function, no shunts, no aortic or tricuspid insufficiency. Biopsy of the tissue resected showed intimal fibroplasia. A TTE completed 1 year after surgery showed normal valvular function and without any structural abnormalities. The patient had improvement in symptoms and an uneventful year after surgical intervention followed by 24 session of cardiac rehabilitation.

 

 

Discussion

Sinus of Valsalva aneurysm is a dilation of the aortic wall between the aortic valve and the sinotubular junction that is caused by the lack of continuity between the middle layer of the aortic wall and the aortic valve.1 Cases of SVA are rare cardiac anomalies with prevalence of 1% in patients undergoing open-heart surgery.2 Between 65% and 85% of SVA cases originate from the right coronary sinus, 10% to 20% from the noncoronary sinus, and < 5% from the left coronary sinus.3

[embed:render:related:node:106996]

Sinus of Valsalva aneurysm is usually congenital, although cases associated with syphilis, bacterial endocarditis, trauma, Behçet disease, and aortic dissection have been reported. Structural defects associated with congenital SVAs include ventricular septal defect, bicuspid aortic valve, and aortic regurgitation. It is less commonly associated with pulmonary stenosis, coarctation of the aorta, patent ductus arteriosus, tricuspid regurgitation, and atrial septal defects.

The most common complication of the SVA is rupture into another cardiac chamber, frequently the right ventricle (60%) or RA (29%) and less frequently into left atrium (6%), left ventricle (4%), or pericardium (1%).1 Patients with ruptured SVA mainly develop dyspnea and chest pain, but cough, fatigue, peripheral edema, and continuous murmur have been reported.1

Atrial septal aneurysm is an uncommon finding in adults, with an incidence of 2.2 % in the general population, and it is often associated with atrial septal defect and PFO.1,4 Although ASA formation can be secondary to interatrial differences in pressures, it can be a primary malformation involving the region of the fossa ovalis or the entire atrial septum.4 Atrial septal aneurysm may be an isolated anomaly, but often is found in association with other structural cardiac anomalies, including SVA and PFO.4,5

Conclusion

Although coexistence of SVA and ASA has been reported previously, the case reported here, a ruptured noncoronary SVA that was associated with a large ASA and a PFO, has not been previously documented in the English literature. This patient’s anomalies are most likely congenital in origin. Progressive dyspnea and chest pain in the presence of a continuous loud murmur should raise the suspicion of ruptured sinus of Valsalva. Although no significant aortic regurgitation was noted on echocardiography, the pistol shot sound heard over the femoral artery was believed to be due to the rapid diastolic runoff into the RA through the ruptured SVA.

The significant increase in the RA pressure made the ASA and PFO more prominent. A TEE, left and right heart catheterizations with shunt study are vital for the diagnosis of SVA. If left untreated, SVA has an ominous prognosis. Surgical repair of ruptured SVA has an accepted risk and good prognosis with 10-year survival rate of 90%, whereas the mean survival of untreated ruptured SVA is about 4 years.6,7 Hence, the patient in this study was referred to a tertiary care center for surgical intervention.

[embed:render:related:node:134882]

References

1. Galicia-Tornell MM, Marín-Solís B, Mercado-Astorga O, Espinoza-Anguiano S, Martínez-Martínez M, Villalpando-Mendoza E. Sinus of Valsalva aneurysm with rupture. Case report and literature review. Cir Cir. 2009;77(6):441-445.

2. Takach TJ, Reul GJ, Duncan JM, et al. Sinus of Valsalva aneurysm or fistula: management and outcome. Ann Thorac Surg. 1999;68(5):1573-1577.

3. Meier JH, Seward JB, Miller FA Jr, Oh JK, Enriquez-Sarano M. Aneurysms in the left ventricular outflow tract: clinical presentation, causes, and echocardiographic features. J Am Soc Echocardiogr. 1998;11(7):729-745.

4. Mügge A, Daniel WG, Angermann C et al. Atrial septal aneurysm in adult patients: a multicenter study using transthoracic and transesophageal echocardiography. Circulation. 1995;91(11):2785-2792.

5. Silver MD, Dorsey JS. Aneurysms of the septum primum in adults. Arch Pathol Lab Med. 1978;102(2):62-65.

6. Wang ZJ, Zou CW, Li DC, et al. Surgical repair of sinus of Valsalva aneurysm in Asian patients. Ann Thorac Surg. 2007;84(1):156-160.

7. Yan F, Huo Q, Qiao J, Murat V, Ma SF. Surgery for sinus of valsalva aneurysm: 27-year experience with 100 patients. Asian Cardiovasc Thorac Ann. 2008;16(5):361-365.

Article PDF
Author and Disclosure Information

Dr. Khattak is a cardiologist at Kettering Medical Center. Dr. Patel is an internal medicine resident and Dr. Al-Zubaidi is cardiology fellow, both at Wright State University. Dr. Tivakaran is a cardiologist at Dayton VAMC; all located in Dayton, Ohio.

Author disclosures
The authors report no actual or potential conflicts of interest 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. 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.

Issue
Federal Practitioner - 34(6)
Publications
Topics
Page Number
33-35
Sections
Author and Disclosure Information

Dr. Khattak is a cardiologist at Kettering Medical Center. Dr. Patel is an internal medicine resident and Dr. Al-Zubaidi is cardiology fellow, both at Wright State University. Dr. Tivakaran is a cardiologist at Dayton VAMC; all located in Dayton, Ohio.

Author disclosures
The authors report no actual or potential conflicts of interest 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. 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.

Author and Disclosure Information

Dr. Khattak is a cardiologist at Kettering Medical Center. Dr. Patel is an internal medicine resident and Dr. Al-Zubaidi is cardiology fellow, both at Wright State University. Dr. Tivakaran is a cardiologist at Dayton VAMC; all located in Dayton, Ohio.

Author disclosures
The authors report no actual or potential conflicts of interest 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. 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.

Article PDF
Article PDF
Related Articles
The confluence of atrial septal aneurysm and patent foramen ovale in noncoronary sinus of Valsalva has not been previously documented in the literature.
The confluence of atrial septal aneurysm and patent foramen ovale in noncoronary sinus of Valsalva has not been previously documented in the literature.

A 53 year-old white male with a past medical history of hypertension, hyperlipidemia, and former tobacco use was referred to the Dayton VAMC in Ohio for symptoms that included shortness of breath and a recent abnormal stress test. The patient reported no history of known coronary artery disease (CAD), congestive heart failure, or other cardiovascular diseases. The patient also reported no recent fever, bacterial blood infection, syphilis infection, recreational drug use, or chest trauma.

A physical examination was remarkable for grade 3/6 continuous murmur at the 5th interspace to the left of the sternum and a loud “pistol shot” sound heard over the femoral artery. The patient had jugular venous distension and 2+ leg edema bilaterally. His vital signs were normal, and laboratory blood tests showed normal hemoglobin level and kidney function.

An electrocardiogram showed nonspecific ST segment changes and a transthoracic echocardiogram (TTE) revealed a high-velocity jet in the right atrium (RA) above the tricuspid valve concerning for sinus of Valsalva aneurysm (SVA).

fed03406033_f1.png
A transesophageal echocardiogram (TEE) showed a “windsock” appearance of the noncoronary SVA with possible rupture into the RA (Figure 1) and atrial septal aneurysm (ASA) with more than 2-cm displacement beyond the plane of the atrial septum and a 2-mm patent foramen ovale (PFO) (Figure 2).
fed03406033_f2.png
fed03406033_f3.png

 

Right heart catheterization revealed elevated RA pressures with positive shunt study showing oxygen saturation step-up in the RA (Figure 3). Left heart hemodynamic measurement from an aortic approach to the distal part of the noncoronary cusp SVA revealed an RA pressure-tracing pattern consistent with rupture of the noncoronary SVA into the RA (Figure 4).

fed03406033_f4.png
Coronary angiography revealed single vessel CAD involving the proximal right coronary artery.

The primary diagnosis was of acute heart failure secondary to ruptured aneurysm of the noncoronary SVA into RA. The patient also received a secondary diagnosis of atrial septal aneurysm and PFO.

[embed:render:related:node:128884]

Treatment & Outcome

The patient was treated with aggressive diuresis and responded well to therapy. Considering the high mortality rate associated with a ruptured SVA, the patient was referred to a tertiary care center for surgical evaluation. He underwent repair of aorto-right atrial communication with a Cormatrix patch (Roswell, GA) from the aortic side and with primary closure from the right atrial side with resection of the windsock tract; coronary artery bypass graft x1 with right internal mammary artery to the right coronary artery; closure of the PFO with the Cormatrix patch.

The postoperative TEE confirmed preserved LV and RV function, no shunts, no aortic or tricuspid insufficiency. Biopsy of the tissue resected showed intimal fibroplasia. A TTE completed 1 year after surgery showed normal valvular function and without any structural abnormalities. The patient had improvement in symptoms and an uneventful year after surgical intervention followed by 24 session of cardiac rehabilitation.

 

 

Discussion

Sinus of Valsalva aneurysm is a dilation of the aortic wall between the aortic valve and the sinotubular junction that is caused by the lack of continuity between the middle layer of the aortic wall and the aortic valve.1 Cases of SVA are rare cardiac anomalies with prevalence of 1% in patients undergoing open-heart surgery.2 Between 65% and 85% of SVA cases originate from the right coronary sinus, 10% to 20% from the noncoronary sinus, and < 5% from the left coronary sinus.3

[embed:render:related:node:106996]

Sinus of Valsalva aneurysm is usually congenital, although cases associated with syphilis, bacterial endocarditis, trauma, Behçet disease, and aortic dissection have been reported. Structural defects associated with congenital SVAs include ventricular septal defect, bicuspid aortic valve, and aortic regurgitation. It is less commonly associated with pulmonary stenosis, coarctation of the aorta, patent ductus arteriosus, tricuspid regurgitation, and atrial septal defects.

The most common complication of the SVA is rupture into another cardiac chamber, frequently the right ventricle (60%) or RA (29%) and less frequently into left atrium (6%), left ventricle (4%), or pericardium (1%).1 Patients with ruptured SVA mainly develop dyspnea and chest pain, but cough, fatigue, peripheral edema, and continuous murmur have been reported.1

Atrial septal aneurysm is an uncommon finding in adults, with an incidence of 2.2 % in the general population, and it is often associated with atrial septal defect and PFO.1,4 Although ASA formation can be secondary to interatrial differences in pressures, it can be a primary malformation involving the region of the fossa ovalis or the entire atrial septum.4 Atrial septal aneurysm may be an isolated anomaly, but often is found in association with other structural cardiac anomalies, including SVA and PFO.4,5

Conclusion

Although coexistence of SVA and ASA has been reported previously, the case reported here, a ruptured noncoronary SVA that was associated with a large ASA and a PFO, has not been previously documented in the English literature. This patient’s anomalies are most likely congenital in origin. Progressive dyspnea and chest pain in the presence of a continuous loud murmur should raise the suspicion of ruptured sinus of Valsalva. Although no significant aortic regurgitation was noted on echocardiography, the pistol shot sound heard over the femoral artery was believed to be due to the rapid diastolic runoff into the RA through the ruptured SVA.

The significant increase in the RA pressure made the ASA and PFO more prominent. A TEE, left and right heart catheterizations with shunt study are vital for the diagnosis of SVA. If left untreated, SVA has an ominous prognosis. Surgical repair of ruptured SVA has an accepted risk and good prognosis with 10-year survival rate of 90%, whereas the mean survival of untreated ruptured SVA is about 4 years.6,7 Hence, the patient in this study was referred to a tertiary care center for surgical intervention.

[embed:render:related:node:134882]

A 53 year-old white male with a past medical history of hypertension, hyperlipidemia, and former tobacco use was referred to the Dayton VAMC in Ohio for symptoms that included shortness of breath and a recent abnormal stress test. The patient reported no history of known coronary artery disease (CAD), congestive heart failure, or other cardiovascular diseases. The patient also reported no recent fever, bacterial blood infection, syphilis infection, recreational drug use, or chest trauma.

A physical examination was remarkable for grade 3/6 continuous murmur at the 5th interspace to the left of the sternum and a loud “pistol shot” sound heard over the femoral artery. The patient had jugular venous distension and 2+ leg edema bilaterally. His vital signs were normal, and laboratory blood tests showed normal hemoglobin level and kidney function.

An electrocardiogram showed nonspecific ST segment changes and a transthoracic echocardiogram (TTE) revealed a high-velocity jet in the right atrium (RA) above the tricuspid valve concerning for sinus of Valsalva aneurysm (SVA).

fed03406033_f1.png
A transesophageal echocardiogram (TEE) showed a “windsock” appearance of the noncoronary SVA with possible rupture into the RA (Figure 1) and atrial septal aneurysm (ASA) with more than 2-cm displacement beyond the plane of the atrial septum and a 2-mm patent foramen ovale (PFO) (Figure 2).
fed03406033_f2.png
fed03406033_f3.png

 

Right heart catheterization revealed elevated RA pressures with positive shunt study showing oxygen saturation step-up in the RA (Figure 3). Left heart hemodynamic measurement from an aortic approach to the distal part of the noncoronary cusp SVA revealed an RA pressure-tracing pattern consistent with rupture of the noncoronary SVA into the RA (Figure 4).

fed03406033_f4.png
Coronary angiography revealed single vessel CAD involving the proximal right coronary artery.

The primary diagnosis was of acute heart failure secondary to ruptured aneurysm of the noncoronary SVA into RA. The patient also received a secondary diagnosis of atrial septal aneurysm and PFO.

[embed:render:related:node:128884]

Treatment & Outcome

The patient was treated with aggressive diuresis and responded well to therapy. Considering the high mortality rate associated with a ruptured SVA, the patient was referred to a tertiary care center for surgical evaluation. He underwent repair of aorto-right atrial communication with a Cormatrix patch (Roswell, GA) from the aortic side and with primary closure from the right atrial side with resection of the windsock tract; coronary artery bypass graft x1 with right internal mammary artery to the right coronary artery; closure of the PFO with the Cormatrix patch.

The postoperative TEE confirmed preserved LV and RV function, no shunts, no aortic or tricuspid insufficiency. Biopsy of the tissue resected showed intimal fibroplasia. A TTE completed 1 year after surgery showed normal valvular function and without any structural abnormalities. The patient had improvement in symptoms and an uneventful year after surgical intervention followed by 24 session of cardiac rehabilitation.

 

 

Discussion

Sinus of Valsalva aneurysm is a dilation of the aortic wall between the aortic valve and the sinotubular junction that is caused by the lack of continuity between the middle layer of the aortic wall and the aortic valve.1 Cases of SVA are rare cardiac anomalies with prevalence of 1% in patients undergoing open-heart surgery.2 Between 65% and 85% of SVA cases originate from the right coronary sinus, 10% to 20% from the noncoronary sinus, and < 5% from the left coronary sinus.3

[embed:render:related:node:106996]

Sinus of Valsalva aneurysm is usually congenital, although cases associated with syphilis, bacterial endocarditis, trauma, Behçet disease, and aortic dissection have been reported. Structural defects associated with congenital SVAs include ventricular septal defect, bicuspid aortic valve, and aortic regurgitation. It is less commonly associated with pulmonary stenosis, coarctation of the aorta, patent ductus arteriosus, tricuspid regurgitation, and atrial septal defects.

The most common complication of the SVA is rupture into another cardiac chamber, frequently the right ventricle (60%) or RA (29%) and less frequently into left atrium (6%), left ventricle (4%), or pericardium (1%).1 Patients with ruptured SVA mainly develop dyspnea and chest pain, but cough, fatigue, peripheral edema, and continuous murmur have been reported.1

Atrial septal aneurysm is an uncommon finding in adults, with an incidence of 2.2 % in the general population, and it is often associated with atrial septal defect and PFO.1,4 Although ASA formation can be secondary to interatrial differences in pressures, it can be a primary malformation involving the region of the fossa ovalis or the entire atrial septum.4 Atrial septal aneurysm may be an isolated anomaly, but often is found in association with other structural cardiac anomalies, including SVA and PFO.4,5

Conclusion

Although coexistence of SVA and ASA has been reported previously, the case reported here, a ruptured noncoronary SVA that was associated with a large ASA and a PFO, has not been previously documented in the English literature. This patient’s anomalies are most likely congenital in origin. Progressive dyspnea and chest pain in the presence of a continuous loud murmur should raise the suspicion of ruptured sinus of Valsalva. Although no significant aortic regurgitation was noted on echocardiography, the pistol shot sound heard over the femoral artery was believed to be due to the rapid diastolic runoff into the RA through the ruptured SVA.

The significant increase in the RA pressure made the ASA and PFO more prominent. A TEE, left and right heart catheterizations with shunt study are vital for the diagnosis of SVA. If left untreated, SVA has an ominous prognosis. Surgical repair of ruptured SVA has an accepted risk and good prognosis with 10-year survival rate of 90%, whereas the mean survival of untreated ruptured SVA is about 4 years.6,7 Hence, the patient in this study was referred to a tertiary care center for surgical intervention.

[embed:render:related:node:134882]

References

1. Galicia-Tornell MM, Marín-Solís B, Mercado-Astorga O, Espinoza-Anguiano S, Martínez-Martínez M, Villalpando-Mendoza E. Sinus of Valsalva aneurysm with rupture. Case report and literature review. Cir Cir. 2009;77(6):441-445.

2. Takach TJ, Reul GJ, Duncan JM, et al. Sinus of Valsalva aneurysm or fistula: management and outcome. Ann Thorac Surg. 1999;68(5):1573-1577.

3. Meier JH, Seward JB, Miller FA Jr, Oh JK, Enriquez-Sarano M. Aneurysms in the left ventricular outflow tract: clinical presentation, causes, and echocardiographic features. J Am Soc Echocardiogr. 1998;11(7):729-745.

4. Mügge A, Daniel WG, Angermann C et al. Atrial septal aneurysm in adult patients: a multicenter study using transthoracic and transesophageal echocardiography. Circulation. 1995;91(11):2785-2792.

5. Silver MD, Dorsey JS. Aneurysms of the septum primum in adults. Arch Pathol Lab Med. 1978;102(2):62-65.

6. Wang ZJ, Zou CW, Li DC, et al. Surgical repair of sinus of Valsalva aneurysm in Asian patients. Ann Thorac Surg. 2007;84(1):156-160.

7. Yan F, Huo Q, Qiao J, Murat V, Ma SF. Surgery for sinus of valsalva aneurysm: 27-year experience with 100 patients. Asian Cardiovasc Thorac Ann. 2008;16(5):361-365.

References

1. Galicia-Tornell MM, Marín-Solís B, Mercado-Astorga O, Espinoza-Anguiano S, Martínez-Martínez M, Villalpando-Mendoza E. Sinus of Valsalva aneurysm with rupture. Case report and literature review. Cir Cir. 2009;77(6):441-445.

2. Takach TJ, Reul GJ, Duncan JM, et al. Sinus of Valsalva aneurysm or fistula: management and outcome. Ann Thorac Surg. 1999;68(5):1573-1577.

3. Meier JH, Seward JB, Miller FA Jr, Oh JK, Enriquez-Sarano M. Aneurysms in the left ventricular outflow tract: clinical presentation, causes, and echocardiographic features. J Am Soc Echocardiogr. 1998;11(7):729-745.

4. Mügge A, Daniel WG, Angermann C et al. Atrial septal aneurysm in adult patients: a multicenter study using transthoracic and transesophageal echocardiography. Circulation. 1995;91(11):2785-2792.

5. Silver MD, Dorsey JS. Aneurysms of the septum primum in adults. Arch Pathol Lab Med. 1978;102(2):62-65.

6. Wang ZJ, Zou CW, Li DC, et al. Surgical repair of sinus of Valsalva aneurysm in Asian patients. Ann Thorac Surg. 2007;84(1):156-160.

7. Yan F, Huo Q, Qiao J, Murat V, Ma SF. Surgery for sinus of valsalva aneurysm: 27-year experience with 100 patients. Asian Cardiovasc Thorac Ann. 2008;16(5):361-365.

Issue
Federal Practitioner - 34(6)
Issue
Federal Practitioner - 34(6)
Page Number
33-35
Page Number
33-35
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Use ProPublica
Article PDF Media
Image
Disable zoom
Off
Media Folder

Periorbital Changes Induced by Prostaglandin Eye Drops

Article Type
Changed
Mon, 04/29/2024 - 11:56
Display Headline
Periorbital Changes Induced by Prostaglandin Eye Drops

To the Editor:

A 42-year man presented with hollowing of the upper eyelid and skin discoloration of the left periorbital area of 10 years’ duration. He was a professional mixed martial arts fighter with a history of 2 surgeries for retinal detachment of the left eye 13 years prior to the current presentation. The patient also has macular scarring in the left eye. He denied a history of facial fracture, reconstructive surgery, or other medical conditions. His visual acuity was unknown; however, he did not require corrective glasses. He used 3 prescription ophthalmic eye drops—dorzolamide hydrochloride plus timolol maleate, 10 mL; brimonidine tartrate ophthalmic solution 0.15%, 5 mL; and latanoprost ophthalmic solution 0.005%, 125 μg/2.5 mL—in the left eye to lower intraocular pressure, as therapy for glaucoma. If left untreated, glaucoma can lead to vision loss.

Physical examination revealed periorbital hyperpigmentation on the left side; hypertrichosis and eyelash trichomegaly compared to the right side; and a deep left upper orbital sulcus compared to the right side (Figure). The patient was alert and oriented to person, place, and time. Extraocular movement was intact bilaterally, and his pupillary reflex was symmetric. No tenderness was noted over the affected area on palpation; no subcutaneous masses or lesions were observed or palpated. There was no ocular discharge, the conjunctiva was pink, and the sclera was white bilaterally.

CT113004025_fig.jpg
%3Cp%3EPeriorbital%20hyperpigmentation%20on%20the%20left%20side%3B%20hypertrichosis%20and%20eyelash%20trichomegaly%20compared%20to%20the%20right%20side%3B%20and%20a%20deep%20left%20upper%20orbital%20sulcus%20compared%20to%20the%20right%20side%20that%20was%20determined%20to%20be%20the%20result%20of%20use%20of%20latanoprost%20ophthalmic%20solution%200.005%25.%3C%2Fp%3E

The differential diagnosis included professional trauma-induced orbital changes, nevus of Ota (oculomucodermal melanocytosis), prostaglandin-associated periorbitopathy (PAP), and melasma. Although the patient sustained an injury that caused retinal detachment, he never experienced an orbital bone fracture; additionally, a fracture would not explain the skin discoloration or longer eyelashes. Periorbital nevus of Ota most commonly manifests as a unilateral scleral and brown-bluish skin discoloration but does not cause hollowing of the orbital sulcus or affect the length and thickness of eyelashes. Melasma—bilateral skin hyperpigmentation that most commonly affects women—can be induced by oral contraceptives, antibiotics, heat, sun exposure, and pregnancy. It does not affect the color of the iris or the depth of the scleral sulcus, and it does not increase the length and thickness of eyelashes. Based on the clinical presentation and a review of the eye drops used, he was diagnosed with PAP due to prolonged use of latanoprost ophthalmic solution. The patient was referred to an ophthalmologist for consideration of a switch to a different class of medication.

Of the 3 eye drops used by this patient, latanoprost, a prostaglandin analog, decreases intraocular pressure and is known to cause PAP. This condition comprises a constellation of changes, including upper eyelid ptosis, deepening of the upper eyelid sulcus, involution of dermatochalasis, periorbital fat atrophy, mild enophthalmos (sunken eye), inferior scleral show, increased prominence of eyelid vessels, and tight eyelids.1 Latanoprost most often produces these findings, but all prostaglandin ophthalmic medications can, including the dual-indication bimatoprost, which was approved by the US Food and Drug Administration to reduce elevated intraocular pressure in patients with open-angle glaucoma or ocular hypertension but also is used to grow darker, thicker, and longer eyelashes. Clinicians who prescribe bimatoprost for this cosmetic indication should be mindful of the potential for PAP and discuss it with patients.

The prescribing information (PI) for bimatoprost (Latisse; Allergan) does not list PAP as an adverse reaction observed in the 4-month multicenter, double-blind, randomized, vehicle-controlled study of bimatoprost (as Latisse) in 278 adults.2 The PI does list “periorbital and lid changes associated with periorbital fat atrophy and skin tightness resulting in deepening of eyelid sulcus and eyelid ptosis” as an adverse reaction in postmarketing experience. However, according to the PI, the frequency of these adverse reactions cannot be established, as the reporting of such incidents was voluntary and the size of the treated population was uncertain.2

Prostaglandins can cause periorbitopathy by several mechanisms; one speculated cause is that this group of medications might provoke smooth muscle contraction. Prostaglandin medications also have an affinity for fat cells1; atrophy of fat cells can lead to enophthalmos and deepening upper eyelid sulcus. In an observational study of 105 participants who were using a prostaglandin in 1 eye for longer than 1 month (the other eye was used as a control), the overall frequency of prostaglandin-associated periorbitopathy was 93.3% in the bimatoprost group, 41.4% in the latanoprost group, and 70% in the travoprost group, while the frequency of deepening of the upper eyelid sulcus was 80% in the bimatoprost group, 15.7% in the latanoprost group, and 45% in the travoprost group.3 These changes may not be as striking when a patient is using a prostaglandin ophthalmic medication in both eyes and may not be noticed even by the patient. It is prudent for the clinician to take a baseline photograph of the patient when these medications are prescribed to observe for early signs of periorbitopathy. These adverse effects may not be reversible when the medication is discontinued4 and have been observed as early as 4 to 6 weeks after the start of treatment.5

Our patient was counseled that his constellation of PAP findings potentially could be partially reversed over months or even a year or longer if the offending agent was discontinued. However, he was cautioned that cessation of latanoprost first needed to be discussed with his ophthalmologist, who would determine if there was a suitable alternative to a prostaglandin analog for him. The patient’s only concern was the aesthetic appearance of the left periorbital area. A hyaluronic acid filler or fat grafting can be considered for correction of orbital sulcus hollowing; however, we could not locate any long-term studies in which such corrective treatments were applied for PAP. Our patient continues to use latanoprost with no change in the frequency of use. There have been no further changes or progression in the physical appearance of the left eye or periorbital area. The patient has not undergone any corrective treatments.

References
  1. Berke SJ. PAP: new concerns for prostaglandin use. Rev Ophthalmol. 2012;19:70.
  2. Latisse (bimatoprost ophthalmic solution 0.03%). Package insert. Allergan; 2021. Accessed April 11, 2024. https://www.rxabbvie.com/pdf/latisse_pi.pdf
  3. Kucukevcilioglu M, Bayer A, Uysal Y, et al. Prostaglandin associated periorbitopathy in patients using bimatoprost, latanoprost and travoprost. Clin Exp Ophthalmol. 2014;42:126-131. doi:10.1111/ceo.12163
  4. Filippopoulos T, Paula JS, Torun N, et al. Periorbital changes associated with topical bimatoprost. Ophthalmic Plast Reconstr Surg. 2008;24:302-307. doi:10.1097/IOP.0b013e31817d81df
  5. Peplinski LS, Smith KA. Deepening of lid sulcus from topical bimatoprost therapy. Optom Vis Sci. 2004;81:574-577. doi:10.1097/01.opx.0000141791.16683.4a
Article PDF
Author and Disclosure Information

Anya Stassiy is from High Point Medspa, Mountainside, New Jersey. Dr. Khachemoune is from SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, Brooklyn, New York.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209 (amorkh@gmail.com).

Publications
Topics
Page Number
E25-E26
Sections
Author and Disclosure Information

Anya Stassiy is from High Point Medspa, Mountainside, New Jersey. Dr. Khachemoune is from SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, Brooklyn, New York.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209 (amorkh@gmail.com).

Author and Disclosure Information

Anya Stassiy is from High Point Medspa, Mountainside, New Jersey. Dr. Khachemoune is from SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, Brooklyn, New York.

The authors report no conflict of interest.

Correspondence: Amor Khachemoune, MD, SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209 (amorkh@gmail.com).

Article PDF
Article PDF

To the Editor:

A 42-year man presented with hollowing of the upper eyelid and skin discoloration of the left periorbital area of 10 years’ duration. He was a professional mixed martial arts fighter with a history of 2 surgeries for retinal detachment of the left eye 13 years prior to the current presentation. The patient also has macular scarring in the left eye. He denied a history of facial fracture, reconstructive surgery, or other medical conditions. His visual acuity was unknown; however, he did not require corrective glasses. He used 3 prescription ophthalmic eye drops—dorzolamide hydrochloride plus timolol maleate, 10 mL; brimonidine tartrate ophthalmic solution 0.15%, 5 mL; and latanoprost ophthalmic solution 0.005%, 125 μg/2.5 mL—in the left eye to lower intraocular pressure, as therapy for glaucoma. If left untreated, glaucoma can lead to vision loss.

Physical examination revealed periorbital hyperpigmentation on the left side; hypertrichosis and eyelash trichomegaly compared to the right side; and a deep left upper orbital sulcus compared to the right side (Figure). The patient was alert and oriented to person, place, and time. Extraocular movement was intact bilaterally, and his pupillary reflex was symmetric. No tenderness was noted over the affected area on palpation; no subcutaneous masses or lesions were observed or palpated. There was no ocular discharge, the conjunctiva was pink, and the sclera was white bilaterally.

CT113004025_fig.jpg
%3Cp%3EPeriorbital%20hyperpigmentation%20on%20the%20left%20side%3B%20hypertrichosis%20and%20eyelash%20trichomegaly%20compared%20to%20the%20right%20side%3B%20and%20a%20deep%20left%20upper%20orbital%20sulcus%20compared%20to%20the%20right%20side%20that%20was%20determined%20to%20be%20the%20result%20of%20use%20of%20latanoprost%20ophthalmic%20solution%200.005%25.%3C%2Fp%3E

The differential diagnosis included professional trauma-induced orbital changes, nevus of Ota (oculomucodermal melanocytosis), prostaglandin-associated periorbitopathy (PAP), and melasma. Although the patient sustained an injury that caused retinal detachment, he never experienced an orbital bone fracture; additionally, a fracture would not explain the skin discoloration or longer eyelashes. Periorbital nevus of Ota most commonly manifests as a unilateral scleral and brown-bluish skin discoloration but does not cause hollowing of the orbital sulcus or affect the length and thickness of eyelashes. Melasma—bilateral skin hyperpigmentation that most commonly affects women—can be induced by oral contraceptives, antibiotics, heat, sun exposure, and pregnancy. It does not affect the color of the iris or the depth of the scleral sulcus, and it does not increase the length and thickness of eyelashes. Based on the clinical presentation and a review of the eye drops used, he was diagnosed with PAP due to prolonged use of latanoprost ophthalmic solution. The patient was referred to an ophthalmologist for consideration of a switch to a different class of medication.

Of the 3 eye drops used by this patient, latanoprost, a prostaglandin analog, decreases intraocular pressure and is known to cause PAP. This condition comprises a constellation of changes, including upper eyelid ptosis, deepening of the upper eyelid sulcus, involution of dermatochalasis, periorbital fat atrophy, mild enophthalmos (sunken eye), inferior scleral show, increased prominence of eyelid vessels, and tight eyelids.1 Latanoprost most often produces these findings, but all prostaglandin ophthalmic medications can, including the dual-indication bimatoprost, which was approved by the US Food and Drug Administration to reduce elevated intraocular pressure in patients with open-angle glaucoma or ocular hypertension but also is used to grow darker, thicker, and longer eyelashes. Clinicians who prescribe bimatoprost for this cosmetic indication should be mindful of the potential for PAP and discuss it with patients.

The prescribing information (PI) for bimatoprost (Latisse; Allergan) does not list PAP as an adverse reaction observed in the 4-month multicenter, double-blind, randomized, vehicle-controlled study of bimatoprost (as Latisse) in 278 adults.2 The PI does list “periorbital and lid changes associated with periorbital fat atrophy and skin tightness resulting in deepening of eyelid sulcus and eyelid ptosis” as an adverse reaction in postmarketing experience. However, according to the PI, the frequency of these adverse reactions cannot be established, as the reporting of such incidents was voluntary and the size of the treated population was uncertain.2

Prostaglandins can cause periorbitopathy by several mechanisms; one speculated cause is that this group of medications might provoke smooth muscle contraction. Prostaglandin medications also have an affinity for fat cells1; atrophy of fat cells can lead to enophthalmos and deepening upper eyelid sulcus. In an observational study of 105 participants who were using a prostaglandin in 1 eye for longer than 1 month (the other eye was used as a control), the overall frequency of prostaglandin-associated periorbitopathy was 93.3% in the bimatoprost group, 41.4% in the latanoprost group, and 70% in the travoprost group, while the frequency of deepening of the upper eyelid sulcus was 80% in the bimatoprost group, 15.7% in the latanoprost group, and 45% in the travoprost group.3 These changes may not be as striking when a patient is using a prostaglandin ophthalmic medication in both eyes and may not be noticed even by the patient. It is prudent for the clinician to take a baseline photograph of the patient when these medications are prescribed to observe for early signs of periorbitopathy. These adverse effects may not be reversible when the medication is discontinued4 and have been observed as early as 4 to 6 weeks after the start of treatment.5

Our patient was counseled that his constellation of PAP findings potentially could be partially reversed over months or even a year or longer if the offending agent was discontinued. However, he was cautioned that cessation of latanoprost first needed to be discussed with his ophthalmologist, who would determine if there was a suitable alternative to a prostaglandin analog for him. The patient’s only concern was the aesthetic appearance of the left periorbital area. A hyaluronic acid filler or fat grafting can be considered for correction of orbital sulcus hollowing; however, we could not locate any long-term studies in which such corrective treatments were applied for PAP. Our patient continues to use latanoprost with no change in the frequency of use. There have been no further changes or progression in the physical appearance of the left eye or periorbital area. The patient has not undergone any corrective treatments.

To the Editor:

A 42-year man presented with hollowing of the upper eyelid and skin discoloration of the left periorbital area of 10 years’ duration. He was a professional mixed martial arts fighter with a history of 2 surgeries for retinal detachment of the left eye 13 years prior to the current presentation. The patient also has macular scarring in the left eye. He denied a history of facial fracture, reconstructive surgery, or other medical conditions. His visual acuity was unknown; however, he did not require corrective glasses. He used 3 prescription ophthalmic eye drops—dorzolamide hydrochloride plus timolol maleate, 10 mL; brimonidine tartrate ophthalmic solution 0.15%, 5 mL; and latanoprost ophthalmic solution 0.005%, 125 μg/2.5 mL—in the left eye to lower intraocular pressure, as therapy for glaucoma. If left untreated, glaucoma can lead to vision loss.

Physical examination revealed periorbital hyperpigmentation on the left side; hypertrichosis and eyelash trichomegaly compared to the right side; and a deep left upper orbital sulcus compared to the right side (Figure). The patient was alert and oriented to person, place, and time. Extraocular movement was intact bilaterally, and his pupillary reflex was symmetric. No tenderness was noted over the affected area on palpation; no subcutaneous masses or lesions were observed or palpated. There was no ocular discharge, the conjunctiva was pink, and the sclera was white bilaterally.

CT113004025_fig.jpg
%3Cp%3EPeriorbital%20hyperpigmentation%20on%20the%20left%20side%3B%20hypertrichosis%20and%20eyelash%20trichomegaly%20compared%20to%20the%20right%20side%3B%20and%20a%20deep%20left%20upper%20orbital%20sulcus%20compared%20to%20the%20right%20side%20that%20was%20determined%20to%20be%20the%20result%20of%20use%20of%20latanoprost%20ophthalmic%20solution%200.005%25.%3C%2Fp%3E

The differential diagnosis included professional trauma-induced orbital changes, nevus of Ota (oculomucodermal melanocytosis), prostaglandin-associated periorbitopathy (PAP), and melasma. Although the patient sustained an injury that caused retinal detachment, he never experienced an orbital bone fracture; additionally, a fracture would not explain the skin discoloration or longer eyelashes. Periorbital nevus of Ota most commonly manifests as a unilateral scleral and brown-bluish skin discoloration but does not cause hollowing of the orbital sulcus or affect the length and thickness of eyelashes. Melasma—bilateral skin hyperpigmentation that most commonly affects women—can be induced by oral contraceptives, antibiotics, heat, sun exposure, and pregnancy. It does not affect the color of the iris or the depth of the scleral sulcus, and it does not increase the length and thickness of eyelashes. Based on the clinical presentation and a review of the eye drops used, he was diagnosed with PAP due to prolonged use of latanoprost ophthalmic solution. The patient was referred to an ophthalmologist for consideration of a switch to a different class of medication.

Of the 3 eye drops used by this patient, latanoprost, a prostaglandin analog, decreases intraocular pressure and is known to cause PAP. This condition comprises a constellation of changes, including upper eyelid ptosis, deepening of the upper eyelid sulcus, involution of dermatochalasis, periorbital fat atrophy, mild enophthalmos (sunken eye), inferior scleral show, increased prominence of eyelid vessels, and tight eyelids.1 Latanoprost most often produces these findings, but all prostaglandin ophthalmic medications can, including the dual-indication bimatoprost, which was approved by the US Food and Drug Administration to reduce elevated intraocular pressure in patients with open-angle glaucoma or ocular hypertension but also is used to grow darker, thicker, and longer eyelashes. Clinicians who prescribe bimatoprost for this cosmetic indication should be mindful of the potential for PAP and discuss it with patients.

The prescribing information (PI) for bimatoprost (Latisse; Allergan) does not list PAP as an adverse reaction observed in the 4-month multicenter, double-blind, randomized, vehicle-controlled study of bimatoprost (as Latisse) in 278 adults.2 The PI does list “periorbital and lid changes associated with periorbital fat atrophy and skin tightness resulting in deepening of eyelid sulcus and eyelid ptosis” as an adverse reaction in postmarketing experience. However, according to the PI, the frequency of these adverse reactions cannot be established, as the reporting of such incidents was voluntary and the size of the treated population was uncertain.2

Prostaglandins can cause periorbitopathy by several mechanisms; one speculated cause is that this group of medications might provoke smooth muscle contraction. Prostaglandin medications also have an affinity for fat cells1; atrophy of fat cells can lead to enophthalmos and deepening upper eyelid sulcus. In an observational study of 105 participants who were using a prostaglandin in 1 eye for longer than 1 month (the other eye was used as a control), the overall frequency of prostaglandin-associated periorbitopathy was 93.3% in the bimatoprost group, 41.4% in the latanoprost group, and 70% in the travoprost group, while the frequency of deepening of the upper eyelid sulcus was 80% in the bimatoprost group, 15.7% in the latanoprost group, and 45% in the travoprost group.3 These changes may not be as striking when a patient is using a prostaglandin ophthalmic medication in both eyes and may not be noticed even by the patient. It is prudent for the clinician to take a baseline photograph of the patient when these medications are prescribed to observe for early signs of periorbitopathy. These adverse effects may not be reversible when the medication is discontinued4 and have been observed as early as 4 to 6 weeks after the start of treatment.5

Our patient was counseled that his constellation of PAP findings potentially could be partially reversed over months or even a year or longer if the offending agent was discontinued. However, he was cautioned that cessation of latanoprost first needed to be discussed with his ophthalmologist, who would determine if there was a suitable alternative to a prostaglandin analog for him. The patient’s only concern was the aesthetic appearance of the left periorbital area. A hyaluronic acid filler or fat grafting can be considered for correction of orbital sulcus hollowing; however, we could not locate any long-term studies in which such corrective treatments were applied for PAP. Our patient continues to use latanoprost with no change in the frequency of use. There have been no further changes or progression in the physical appearance of the left eye or periorbital area. The patient has not undergone any corrective treatments.

References
  1. Berke SJ. PAP: new concerns for prostaglandin use. Rev Ophthalmol. 2012;19:70.
  2. Latisse (bimatoprost ophthalmic solution 0.03%). Package insert. Allergan; 2021. Accessed April 11, 2024. https://www.rxabbvie.com/pdf/latisse_pi.pdf
  3. Kucukevcilioglu M, Bayer A, Uysal Y, et al. Prostaglandin associated periorbitopathy in patients using bimatoprost, latanoprost and travoprost. Clin Exp Ophthalmol. 2014;42:126-131. doi:10.1111/ceo.12163
  4. Filippopoulos T, Paula JS, Torun N, et al. Periorbital changes associated with topical bimatoprost. Ophthalmic Plast Reconstr Surg. 2008;24:302-307. doi:10.1097/IOP.0b013e31817d81df
  5. Peplinski LS, Smith KA. Deepening of lid sulcus from topical bimatoprost therapy. Optom Vis Sci. 2004;81:574-577. doi:10.1097/01.opx.0000141791.16683.4a
References
  1. Berke SJ. PAP: new concerns for prostaglandin use. Rev Ophthalmol. 2012;19:70.
  2. Latisse (bimatoprost ophthalmic solution 0.03%). Package insert. Allergan; 2021. Accessed April 11, 2024. https://www.rxabbvie.com/pdf/latisse_pi.pdf
  3. Kucukevcilioglu M, Bayer A, Uysal Y, et al. Prostaglandin associated periorbitopathy in patients using bimatoprost, latanoprost and travoprost. Clin Exp Ophthalmol. 2014;42:126-131. doi:10.1111/ceo.12163
  4. Filippopoulos T, Paula JS, Torun N, et al. Periorbital changes associated with topical bimatoprost. Ophthalmic Plast Reconstr Surg. 2008;24:302-307. doi:10.1097/IOP.0b013e31817d81df
  5. Peplinski LS, Smith KA. Deepening of lid sulcus from topical bimatoprost therapy. Optom Vis Sci. 2004;81:574-577. doi:10.1097/01.opx.0000141791.16683.4a
Page Number
E25-E26
Page Number
E25-E26
Publications
Publications
Topics
Article Type
Display Headline
Periorbital Changes Induced by Prostaglandin Eye Drops
Display Headline
Periorbital Changes Induced by Prostaglandin Eye Drops
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>Stassiy</fileName> <TBEID>0C02F6CA.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F6CA</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Periorbital Changes Induced by&#13;</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240429T094019</firstPublished> <LastPublished>20240429T094019</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240429T094019</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Anya Stassiy, PA-C; Amor Khachemoune, MD</byline> <bylineText>Anya Stassiy, PA-C; Amor Khachemoune, MD</bylineText> <bylineFull>Anya Stassiy, PA-C; Amor Khachemoune, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType>(choose one)</newsDocType> <journalDocType>(choose one)</journalDocType> <linkLabel/> <pageRange>E25-E26</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:A 42-year man presented with hollowing of the upper eyelid and skin discoloration of the left periorbital area of 10 years’ duration. He was a pro</metaDescription> <articlePDF>301206</articlePDF> <teaserImage/> <title>Periorbital Changes Induced by Prostaglandin Eye Drops</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>2165</CMSID> </CMSIDs> <keywords> <keyword>pigmentation disorder</keyword> <keyword> prostaglandin eye drops</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 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">45</term> </sections> <topics> <term canonical="true">276</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002718.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Periorbital Changes Induced by Prostaglandin Eye Drops</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>A 42-year man presented with hollowing of the upper eyelid and skin discoloration of the left periorbital area of 10 years’ duration. He was a professional mixed martial arts fighter with a history of 2 surgeries for retinal detachment of the left eye 13 years prior to the current presentation. The patient also has macular scarring in the left eye. He denied a history of facial fracture, reconstructive surgery, or other medical conditions. His visual acuity was unknown; however, he did not require corrective glasses. He used 3 prescription ophthalmic eye drops—dorzolamide hydrochloride plus timolol maleate, 10 mL; brimonidine tartrate ophthalmic solution 0.15%, 5 mL; and latanoprost ophthalmic solution 0.005%, 125 μg/2.5 mL—in the left eye to lower intraocular pressure, as therapy for glaucoma. If left untreated, glaucoma can lead to vision loss.</p> <p>Physical examination revealed periorbital hyperpigmentation on the left side; hypertrichosis and eyelash trichomegaly compared to the right side; and a deep left upper orbital sulcus compared to the right side (Figure). The patient was alert and oriented to person, place, and time. Extraocular movement was intact bilaterally, and his pupillary reflex was symmetric. No tenderness was noted over the affected area on palpation; no subcutaneous masses or lesions were observed or palpated. There was no ocular discharge, the conjunctiva was pink, and the sclera was white bilaterally.<br/><br/>The differential diagnosis included professional trauma-induced orbital changes, nevus of Ota (o<span class="hgkelc">culomucodermal melanocytosis)</span>, prostaglandin-associated periorbitopathy (PAP), and melasma. Although the patient sustained an injury that caused retinal detachment, he never experienced an orbital bone fracture; additionally, a fracture would not explain the skin discoloration or longer eyelashes. Periorbital nevus of Ota most commonly manifests as a unilateral scleral and brown-bluish skin discoloration but does not cause hollowing of the orbital sulcus or affect the length and thickness of eyelashes. Melasma—bilateral skin hyperpigmentation that most commonly affects women—can be induced by oral contraceptives, antibiotics, heat, sun exposure, and pregnancy. It does not affect the color of the iris or the depth of the scleral sulcus, and it does not increase the length and thickness of eyelashes.<b> </b>Based on the clinical presentation and a review of the eye drops used, he was diagnosed with PAP due to prolonged use of latanoprost ophthalmic solution. The patient was referred to an ophthalmologist for consideration of a switch to a different class of medication.<br/><br/>Of the 3 eye drops used by this patient, latanoprost, a prostaglandin analog, decreases intraocular pressure and is known to cause PAP. This condition comprises a constellation of changes, including upper eyelid ptosis, deepening of the upper eyelid sulcus, involution of dermatochalasis, periorbital fat atrophy, mild enophthalmos (sunken eye), inferior scleral show, increased prominence of eyelid vessels, and tight eyelids.<sup>1</sup> Latanoprost most often produces these findings, but all prostaglandin ophthalmic medications can, including the dual-indication bimatoprost, which was approved by the US Food and Drug Administration to reduce elevated intraocular pressure in patients with open-angle glaucoma or ocular hypertension but also is used to grow darker, thicker, and longer eyelashes. Clinicians who prescribe bimatoprost for this cosmetic indication should be mindful of the potential for PAP and discuss it with patients.<br/><br/>The prescribing information (PI) for bimatoprost (Latisse; Allergan) does not list PAP as an adverse reaction observed in the 4-month multicenter, double-blind, randomized, vehicle-controlled study of bimatoprost (as Latisse) in 278 adults.<sup>2</sup> The PI does list “periorbital and lid changes associated with periorbital fat atrophy and skin tightness resulting in deepening of eyelid sulcus and eyelid ptosis” as an adverse reaction in postmarketing experience. However, according to the PI, the frequency of these adverse reactions cannot be established, as the reporting of such incidents was voluntary and the size of the treated population was uncertain.<sup>2<br/><br/></sup>Prostaglandins can cause periorbitopathy by several mechanisms; one speculated cause is that this group of medications might provoke smooth muscle contraction. Prostaglandin medications also have an affinity for fat cells<sup>1</sup>; atrophy of fat cells can lead to enophthalmos and deepening upper eyelid sulcus. In an observational study of 105 participants who were using a prostaglandin in 1 eye for longer than 1 month (the other eye was used as a control), the overall frequency of prostaglandin-associated periorbitopathy was 93.3% in the bimatoprost group, 41.4% in the latanoprost group, and 70% in the travoprost group, while the frequency of deepening of the upper eyelid sulcus was 80% in the bimatoprost group, 15.7% in the latanoprost group, and 45% in the travoprost group.<sup>3</sup> These changes may not be as striking when a patient is using a prostaglandin ophthalmic medication in both eyes and may not be noticed even by the patient. It is prudent for the clinician to take a baseline photograph of the patient when these medications are prescribed to observe for early signs of periorbitopathy. These adverse effects may not be reversible when the medication is discontinued<sup>4</sup> and have been observed as early as 4 to 6 weeks after the start of treatment.<sup>5<br/><br/></sup>Our patient was counseled that his constellation of PAP findings potentially could be partially reversed over months or even a year or longer if the offending agent was discontinued. However, he was cautioned that cessation of latanoprost first needed to be discussed with his ophthalmologist, who would determine if there was a suitable alternative to a prostaglandin analog for him. The patient’s only concern was the aesthetic appearance of the left periorbital area. A hyaluronic acid filler or fat grafting can be considered for correction of orbital sulcus hollowing; however, we could not locate any long-term studies in which such corrective treatments were applied for PAP. Our patient continues to use latanoprost with no change in the frequency of use. There have been no further changes or progression in the physical appearance of the left eye or periorbital area. The patient has not undergone any corrective treatments.</p> <h2>REFERENCES</h2> <p class="reference"> 1. Berke SJ. PAP: new concerns for prostaglandin use. <i>Rev Ophthalmol</i>. 2012;19:70. <br/><br/> 2. Latisse (bimatoprost ophthalmic solution 0.03%). Package insert. Allergan; 2021. Accessed April 11, 2024. https://www.rxabbvie.com/pdf/latisse_pi.pdf<br/><br/> 3. Kucukevcilioglu M, Bayer A, Uysal Y, et al. Prostaglandin associated periorbitopathy in patients using bimatoprost, latanoprost and travoprost<i>. Clin Exp Ophthalmol. </i>2014;42:126-131. doi:10.1111/ceo.12163<br/><br/> 4. Filippopoulos T, Paula JS, Torun N, et al. Periorbital changes associated with topical bimatoprost.<i> Ophthalmic Plast Reconstr Surg</i>. 2008;24:302-307. doi:10.1097/IOP.0b013e31817d81df<br/><br/> 5. Peplinski LS, Smith KA. Deepening of lid sulcus from topical bimatoprost therapy. <span class="Emphasis">Optom Vis Sci.</span><span class="Emphasis"> 2004;</span>81:574-577. <span class="citation-doi">doi:10.1097/01.opx.0000141791.16683.4a</span></p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Anya Stassiy is from High Point Medspa, Mountainside, New Jersey. Dr. Khachemoune is from SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, Brooklyn, New York.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Amor Khachemoune, MD, SUNY Downstate Health Sciences University, Veterans Affairs Medical Center, 800 Poly Pl, Brooklyn, NY 11209 (amorkh@gmail.com).<br/><br/><i>Cutis</i>. 2024 April;113(4):E25-E26. doi:10.12788/cutis.1005</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>fig</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="caption">Periorbital hyperpigmentation on the left side; hypertrichosis and eyelash trichomegaly compared to the right side; and a deep left upper orbital sulcus compared to the right side that was determined to be the result of use of latanoprost ophthalmic solution 0.005%.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>pp</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Ask patients to provide photographs taken prior to noticed changes to assess progression if they are new to your practice.</li> <li>Take photographs of patients in good light against a solid-colored background to have a baseline. It may be helpful to update patient images annually.</li> <li>Discuss with patients the aesthetic changes that may occur with the use of prescription medications. Provide pamphlets with images to educate them on what to expect.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Citation Override
doi:10.12788/cutis.1005
Inside the Article

PRACTICE POINTS

  • Ask patients to provide photographs taken prior to noticed changes to assess progression if they are new to your practice.
  • Take photographs of patients in good light against a solid-colored background to have a baseline. It may be helpful to update patient images annually.
  • Discuss with patients the aesthetic changes that may occur with the use of prescription medications. Provide pamphlets with images to educate them on what to expect.
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

Recurrent Soft Tissue Rosai Dorfman Disease of Right Medial Thigh Lipoma With Lymph Node Involvement

Article Type
Changed
Mon, 04/29/2024 - 19:30

Rosai Dorfman disease (RDD) is a rare non-Langerhans cell histiocytosis first described in 1965 by Destombes and again in 1969 by Rosai and Dorfman to depict patients who presented with massive cervical lymphadenopathy.1 The classification for histiocytosis was revised in 2016 based on new insights into the pathologic, genetic, and molecular features of RDD.2,3 Now, RDD is listed under the R group, which includes familial, sporadic, classical (nodal), extranodal RDD, and other noncutaneous, non-Langerhans cell histiocytosis.3 Cutaneous RDD is classified under the C group and typically presents as painless papules, plaques, or nodules without significant lymphadenopathy, or systemic symptoms usually seen in the presentation of RDD.4

The etiology of RDD is poorly understood, although an underlying infectious or genetic component is suspected.5 Several pathogens—including human herpesvirus 6, parvovirus B19, Epstein-Barr virus, cytomegalovirus, Brucella, and Klebsiella—have all been investigated. A link to kinase mutations has been described in nodal and extranodal RDD; however, the molecular profile of cutaneous RDD remains unknown.2 Histologic findings for RDD typically include cells that are S100 positive, CD68 positive, and CD163 positive, and CD1a and langerin (CD207) negative, thus excluding Langerhans cell histiocytosis.2 The hallmark finding of RDD is emperipolesis, which results from “histiocyte-mediated phagocytosis of intact lymphocytes and other immune cells.”6 Immunoglobulin G (Ig) G4-positive plasma cells are also common, but the significance of this finding is controversial. We present a case of a patient with recurrent RDD within a right medial thigh lipoma and include a literature review to explore the significance of histologic findings and various treatment options in the setting of emerging treatment and diagnostic criteria.

 

Case Presentation

A 56-year-old African American male was evaluated in the rheumatology clinic at the Central Texas Veterans Affairs Medical Center in Temple, Texas, in 2022 for a cutaneous mass of his right medial thigh. The patient previously reported the onset of a right medial thigh mass in 2005 after he had been deployed in Iraq for about 1 year. A biopsy of the mass from 2005 showed infiltration of plasma cells, lymphocytes, and histiocytes and occasional neutrophils with noted reactivity of S100 protein and CD163, but not CD1a. The patient’s original biopsy report from March 2005 was obtained secondhand from an addendum to a Dermatology Consult note. Surgical excision of the mass was not performed until 2012 and systemic therapy was not initiated.

figure_1.png

In 2021, the mass recurred and gradually increased in size, prompting a second surgical removal. Pathology results from the 2021 mass showed a lipoma with areas of fibrosis with a mixed inflammatory cell infiltrate, including abundant lymphocytes, plasma cells, occasional hemosiderin-laden histiocytes, and clusters of enlarged histiocytes with foamy to pale eosinophilic, finely granular cytoplasm, and large, round, vesicular nuclei with prominent nucleoli. Emperipolesis was also present (Figure 1).

figure_2.png

Special immunohistochemical staining showed most of the lymphocytes were CD20 positive B-cells with a minority of CD3 positive T-cells. Histiocytes were CD163 positive and CD68 positive with patchy reactivity for S100 protein. The plasma cells were CD138 positive. There were > 125 IgG4-positive plasma cells present in a single high-powered field and the overall IgG4:IgG plasma cell ratio was > 40%. Pertinent imaging included a whole-body positron emission tomography/computed tomography (PET/CT) hypermetabolic activity scan of a small right femoral lymph node (9 mm) and nearby medial right femoral lymph node (13 mm) (Figure 2A). A well-defined mass in the medial aspect of the right thigh (2.5 cm x 3.2 cm x 3.9 cm) and a cutaneous/subcutaneous lesion of the anterior medial aspect of the proximal right thigh superior to the mass (2.9 cm) were also evident on imaging (Figure 2B). Each area of hypermetabolic activity had decreased in size and activity when compared to a previous PET/CT obtained 1 month earlier. There was no evidence of skeletal malignancy. A physical examination did not reveal any other soft tissue masses, palpable lymphadenopathy, or areas of skin involvement. Given the patient’s reassuring imaging findings and a lack of any new physical examination findings, no systemic therapy was initiated, and following shared decision making, the patient agreed to a period of watchful waiting. 

 

 

Discussion

RDD is rare with a prevalence of 1:200,000. It has been reported that multisystem involvement occurs in 19% of cases and the prognosis of RDD correlates with the number of extranodal systems involved in the disease process.7 Although sporadic RDD is usually self-limited with favorable outcomes, it is estimated that 10% of patients may die of RDD due to direct complications, infections, and amyloidosis.2,7 RDD commonly affects young male children and young adults with a mean age of 20 years and has a higher incidence among African American children.2,7,8 Although patients with RDD present bilateral, painless cervical lymphadenopathy in 90% of cases, about 43% of patients with RDD and associated adenopathy present with ≥ 1 site of extranodal involvement, and only 23% of patients with RDD present with isolated extranodal sites without adenopathy.9 As was the case with our patient, the most common extranodal sites are found in the skin and soft tissue (16%).6,9 However, histopathologic diagnosis of RDD in a lipoma is exceedingly rare. We found only 1 other case report of a patient with a history of multiple lipomas who developed a new solitary nodule that was excised and demonstrated RDD upon immunohistochemical staining.4 There has been no documented association between multiple lipomas and RDD.4

Histologically, RDD is often characterized by emperipolesis (the presence of an intact cell within the cytoplasm of anther cell) and a mixed cell infiltrate that includes S100 positive histiocytes, mononuclear cells, plasma cells, and lymphocytes.10 Despite these shared histologic features among the various phenotypes of RDD, other type-specific characteristics may also be present. When compared to nodal RDD, extranodal disease tends to demonstrate a lack of nodal architecture, more fibrosis and collagen deposition, fewer RDD cells, a lower degree of emperipolesis, and alternating pale (histiocyte rich) and dark (lymphocyte rich) regions with notable polygonal histiocytes arranged in a storiform pattern.5,10

Our patient’s histology showed an overall IgG4:IgG plasma cell ratio > 40%. RDD frequently presents with IgG4-positive plasma cells, which has confounded the diagnosis of IgG4-related diseases and hyper-IgG4 disease.11 Given this association, the Histiocyte Society revised classification recommends that all cases of RDD be evaluated for IgG4-positive cell infiltration.2,3 Further discussion on this matter was recently provided after an expert panel published a consensus statement in 2015 detailing the evaluation of IgG4. The panel advocates for stricter terminology and criteria on this issue, advises that isolated IgG4-positive plasma cells are nonspecific, and states that the diagnosis of IgG4 disease should be based on careful judgment and correlation with the clinical scenario and supportive findings.12 Therefore, while IgG4 positivity continues to be misleading in RDD cases, further evaluation for IgG4 disease is recommended. 

Sporadic RDD is usually self-limited with a reported remission rate of up to 50%, according to a case series of 80 patients with RDD.13 This leads to the recommendation of a period of watchful monitoring in patients with limited disease.13 In patients with unifocal extranodal disease, surgical excision has shown positive remission results; however, local recurrence of soft tissue lesions can occur at a rate of 21.4% to 51%.14 Although initiation of systemic therapy should be considered in patients with recurrent disease, there is currently no standardized regimen or medication of choice for treatment. Treatment with steroids, including prednisone 40 to 70 mg daily or dexamethasone 8 to 20 mg daily, have been shown to be effective in reducing the nodal size and symptoms, especially in cases of nonresectable multifocal extranodal disease of the central nervous system, bone, and orbital.7,15,16 However, cases of orbital, tracheal, renal, or soft tissue RDD have reported failure in treatment with steroids.17,18

According to the consensus recommendations for the treatment of RDD released in 2018, treatment with chemotherapy has shown mixed results. Anthracycline and alkylating agents have shown minimal efficacy, but combination regimens with vinca alkaloids, methotrexate, and 6-mercaptopurine have helped patients experience remission.19,20 Due to the rarity of RDD and lack of clinical trials, the exact efficacy of these treatment regimens remains unknown and is largely limited to case reports described within the medical literature. Treatment with nucleoside analogs, such as cladribine 2.1 to 5 mg/m2 or clofarabine 25 mg/m2 per day for 5 days every 28 days for 6 months, have shown promising results and helped achieve complete remission in patients with refractory or recurrent RDD.7,21-23 Immunomodulator therapies including TNF-α inhibitor, such as thalidomide and lenalidomide, have also shown to be effective, particularly in patients with refractory disease.24,25 Low-dose thalidomide (100 mg daily) was effective for cases of refractory cutaneous RDD, though no standard dosing regimen exists. Lenalidomide has shown to be effective in patients with multiple refractory nodal or bone RDD, but is associated with more complications given that it is more myelosuppressive than thalidomide.7 Radiotherapy has also been initiated in patients with refractory soft tissue disease or persistent symptoms after resection and in patients who are not candidates for surgery or systemic therapy, though no standard doses of radiotherapy have been established.7,26,27

Conclusions

RDD is a rare histiocytic disorder that presents in a wide range of age groups, different locations in the body, and with variable disease behavior. Multidisciplinary management of the disease and research for mutations and microenvironment of RDD is needed to better understand its clinicopathological nature and improve targeted novel therapies.

Acknowledgments

The authors thank Veterans Affairs Central Texas Health Care Section Chief of Rheumatology, Swastika Jha, MD, for her guidance in this case and Bo Wang, MD, for his preparation of the pathological specimens. 

References

1. Goyal G, Ravindran A, Young JR, et al. Clinicopathological features, treatment approaches, and outcomes in Rosai-Dorfman disease. Haematologica. 2020;105(2):348-357. Published 2020 Jan 31. doi:10.3324/haematol.2019.219626

2. Bruce-Brand C, Schneider JW, Schubert P. Rosai-Dorfman disease: an overview. J Clin Pathol. 2020;73(11):697-705. doi:10.1136/jclinpath-2020-206733

3. Emile JF, Abla O, Fraitag S, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. 2016;127(22):2672-2681. doi:10.1182/blood-2016-01-690636

4. Farooq U, Chacon AH, Vincek V, Elgart G. Purely cutaneous rosai-dorfman disease with immunohistochemistry. Indian J Dermatol. 2013;58(6):447-450. doi:10.4103/0019-5154.119953

5. Ma H, Zheng Y, Zhu G, Wu J, Lu C, Lai W. Rosai-dorfman disease with massive cutaneous nodule on the shoulder and back. Ann Dermatol. 2015;27(1):71-75. doi:10.5021/ad.2015.27.1.71

6. Deen IU, Chittal A, Badro N, Jones R, Haas C. Extranodal Rosai-Dorfman Disease- a Review of Diagnostic Testing and Management. J Community Hosp Intern Med Perspect. 2022;12(2):18-22. Published 2022 Apr 12. doi:10.55729/2000-9666.1032

7. Oussama A, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877-2890. doi: 10.1182/blood-2018-03-839753

8. Foucar E, Rosai J, Dorfman R. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): review of the entity. Semin Diagn Pathol. 1990;7(1):19-73.

9. Gaitonde S. Multifocal, extranodal sinus histiocytosis with massive lymphadenopathy: an overview. Arch Pathol Lab Med. 2007;131(7):1111-1121. doi:10.5858/2007-131-1117-MESHWM

10. Betini N, Munger AM, Rottmann D, Haims A, Costa J, Lindskog DM. Rare presentation of Rosai-Dorfman disease in soft tissue: diagnostic findings and surgical treatment. Case Rep Surg. 2022;2022:8440836. Published 2022 Mar 30. doi:10.1155/2022/8440836

11. Menon MP, Evbuomwan MO, Rosai J, Jaffe ES, Pittaluga S. A subset of Rosai-Dorfman disease cases show increased IgG4-positive plasma cells: another red herring or a true association with IgG4-related disease? Histopathology. 2014;64(3):455-459. doi:10.1111/his.12274

12. Khosroshahi A, Wallace ZS, Crowe JL, et al. International consensus guidance statement on the management and treatment of IgG4-related disease. Arthritis Rheumatol. 2015;67(7):1688-1699. doi:10.1002/art.39132

13. Pulsoni A, Anghel G, Falcucci P, et al. Treatment of sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): report of a case and literature review. Am J Hematol. 2002;69(1):67-71. doi:10.1002/ajh.10008

14. Montgomery EA, Meis JM, Firzzera G. Rosai-Dorfman disease of soft tissue. Am J Surg Pathol. 1992;16(2):122-129. doi:10.1097/00000478-199202000-00004

15. Z’Graggen WJ, Sturzenegger M, Mariani L, Keserue B, Kappeler A, Vajtai I. Isolated Rosai-Dorfman disease of intracranial meninges. Pathol Res Pract. 2006;202(3):165-170. doi:10.1016/j.prp.2005.11.004

16. Shulman S, Katzenstein H, Abramowsky C, Broecker J, Wulkan M, Shehata B. Unusual presentation of Rosai-Dorfman disease (RDD) in the bone in adolescents. Fetal Pediatr Pathol. 2011;30(6):442-447. doi:10.3109/15513815.2011.61887317. Ottaviano G, Doro D, Marioni G, et al. Extranodal Rosai-Dorfman disease: involvement of eye, nose and trachea. Acta Otolaryngol. 2006;126(6):657-660. doi:10.1080/00016480500452582

18. Sakallioglu O, Gok F, Kalman S, et al. Minimal change nephropathy in a 7-year-old boy with Rosai-Dorfman disease. J Nephrol. 2006;19(2):211-214.

19. Jabali Y, Smrcka V, Pradna J. Rosai-Dorfman disease: successful long-term results by combination chemotherapy with prednisone, 6-mercaptopurine, methotrexate, and vinblastine: a case report. Int J Surg Pathol. 2005;13(3):285-289. doi:10.1177/106689690501300311

20. Abla O, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877-2890. doi:10.1182/blood-2018-03-839753

21. Konca C, Özkurt ZN, Deger M, Akı Z, Yagcı M. Extranodal multifocal Rosai-Dorfman disease: response to 2-chlorodeoxyadenosine treatment. Int J Hematol. 2009;89(1):58-62. doi:10.1007/s12185-008-0192-2

22. Aouba A, Terrier B, Vasiliu V, et al. Dramatic clinical efficacy of cladribine in Rosai-Dorfman disease and evolution of the cytokine profile: towards a new therapeutic approach. Haematologica. 2006;91(12 Suppl):ECR52.

23. Tasso M, Esquembre C, Blanco E, Moscardó C, Niveiro M, Payá A. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) treated with 2-chlorodeoxyadenosine. Pediatr Blood Cancer. 2006;47(5):612-615. doi:10.1002/pbc.20668

24. Chen E, Pavlidakey P, Sami N. Rosai-Dorfman disease successfully treated with thalidomide. JAAD Case Reports. 2016;2(5):369-372. Published 2016 Sep 28. doi:10.1016/j.jdcr.2016.08.006

25. Rubinstein M, Assal A, Scherba M, et al. Lenalidomide in the treatment of Rosai Dorfman disease-a first in use report. Am J Hematol. 2016;91(2):E1. doi:10.1002/ajh.24225

26. Sandoval-Sus JD, Sandoval-Leon AC, Chapman JR, et al. Rosai-Dorfman disease of the central nervous system: report of 6 cases and review of the literature. Medicine (Baltimore). 2014;93(3):165-175. doi:10.1097/MD.0000000000000030

27. Paryani NN, Daugherty LC, O’Connor MI, Jiang L. Extranodal Rosai-Dorfman disease of the bone treated with surgery and radiotherapy. Rare Tumors. 2014;6(4):5531. Published 2014 Dec 11. doi:10.4081/rt.2014.5531

Article PDF
Author and Disclosure Information

John Corbyn Cravero, MDa; Samar Ibrahim, MDa

Correspondence:  John Cravero  (jccravero13@gmail.com)

aBaylor Scott & White Medical Center – Temple, Texas

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 FederalPractitioner, 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 informed consent for publication was obtained by the patient who was involved in this case report.

Issue
Federal Practitioner - 41(5)s
Publications
Topics
Page Number
S20
Sections
Author and Disclosure Information

John Corbyn Cravero, MDa; Samar Ibrahim, MDa

Correspondence:  John Cravero  (jccravero13@gmail.com)

aBaylor Scott & White Medical Center – Temple, Texas

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 FederalPractitioner, 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 informed consent for publication was obtained by the patient who was involved in this case report.

Author and Disclosure Information

John Corbyn Cravero, MDa; Samar Ibrahim, MDa

Correspondence:  John Cravero  (jccravero13@gmail.com)

aBaylor Scott & White Medical Center – Temple, Texas

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 FederalPractitioner, 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 informed consent for publication was obtained by the patient who was involved in this case report.

Article PDF
Article PDF

Rosai Dorfman disease (RDD) is a rare non-Langerhans cell histiocytosis first described in 1965 by Destombes and again in 1969 by Rosai and Dorfman to depict patients who presented with massive cervical lymphadenopathy.1 The classification for histiocytosis was revised in 2016 based on new insights into the pathologic, genetic, and molecular features of RDD.2,3 Now, RDD is listed under the R group, which includes familial, sporadic, classical (nodal), extranodal RDD, and other noncutaneous, non-Langerhans cell histiocytosis.3 Cutaneous RDD is classified under the C group and typically presents as painless papules, plaques, or nodules without significant lymphadenopathy, or systemic symptoms usually seen in the presentation of RDD.4

The etiology of RDD is poorly understood, although an underlying infectious or genetic component is suspected.5 Several pathogens—including human herpesvirus 6, parvovirus B19, Epstein-Barr virus, cytomegalovirus, Brucella, and Klebsiella—have all been investigated. A link to kinase mutations has been described in nodal and extranodal RDD; however, the molecular profile of cutaneous RDD remains unknown.2 Histologic findings for RDD typically include cells that are S100 positive, CD68 positive, and CD163 positive, and CD1a and langerin (CD207) negative, thus excluding Langerhans cell histiocytosis.2 The hallmark finding of RDD is emperipolesis, which results from “histiocyte-mediated phagocytosis of intact lymphocytes and other immune cells.”6 Immunoglobulin G (Ig) G4-positive plasma cells are also common, but the significance of this finding is controversial. We present a case of a patient with recurrent RDD within a right medial thigh lipoma and include a literature review to explore the significance of histologic findings and various treatment options in the setting of emerging treatment and diagnostic criteria.

 

Case Presentation

A 56-year-old African American male was evaluated in the rheumatology clinic at the Central Texas Veterans Affairs Medical Center in Temple, Texas, in 2022 for a cutaneous mass of his right medial thigh. The patient previously reported the onset of a right medial thigh mass in 2005 after he had been deployed in Iraq for about 1 year. A biopsy of the mass from 2005 showed infiltration of plasma cells, lymphocytes, and histiocytes and occasional neutrophils with noted reactivity of S100 protein and CD163, but not CD1a. The patient’s original biopsy report from March 2005 was obtained secondhand from an addendum to a Dermatology Consult note. Surgical excision of the mass was not performed until 2012 and systemic therapy was not initiated.

figure_1.png

In 2021, the mass recurred and gradually increased in size, prompting a second surgical removal. Pathology results from the 2021 mass showed a lipoma with areas of fibrosis with a mixed inflammatory cell infiltrate, including abundant lymphocytes, plasma cells, occasional hemosiderin-laden histiocytes, and clusters of enlarged histiocytes with foamy to pale eosinophilic, finely granular cytoplasm, and large, round, vesicular nuclei with prominent nucleoli. Emperipolesis was also present (Figure 1).

figure_2.png

Special immunohistochemical staining showed most of the lymphocytes were CD20 positive B-cells with a minority of CD3 positive T-cells. Histiocytes were CD163 positive and CD68 positive with patchy reactivity for S100 protein. The plasma cells were CD138 positive. There were > 125 IgG4-positive plasma cells present in a single high-powered field and the overall IgG4:IgG plasma cell ratio was > 40%. Pertinent imaging included a whole-body positron emission tomography/computed tomography (PET/CT) hypermetabolic activity scan of a small right femoral lymph node (9 mm) and nearby medial right femoral lymph node (13 mm) (Figure 2A). A well-defined mass in the medial aspect of the right thigh (2.5 cm x 3.2 cm x 3.9 cm) and a cutaneous/subcutaneous lesion of the anterior medial aspect of the proximal right thigh superior to the mass (2.9 cm) were also evident on imaging (Figure 2B). Each area of hypermetabolic activity had decreased in size and activity when compared to a previous PET/CT obtained 1 month earlier. There was no evidence of skeletal malignancy. A physical examination did not reveal any other soft tissue masses, palpable lymphadenopathy, or areas of skin involvement. Given the patient’s reassuring imaging findings and a lack of any new physical examination findings, no systemic therapy was initiated, and following shared decision making, the patient agreed to a period of watchful waiting. 

 

 

Discussion

RDD is rare with a prevalence of 1:200,000. It has been reported that multisystem involvement occurs in 19% of cases and the prognosis of RDD correlates with the number of extranodal systems involved in the disease process.7 Although sporadic RDD is usually self-limited with favorable outcomes, it is estimated that 10% of patients may die of RDD due to direct complications, infections, and amyloidosis.2,7 RDD commonly affects young male children and young adults with a mean age of 20 years and has a higher incidence among African American children.2,7,8 Although patients with RDD present bilateral, painless cervical lymphadenopathy in 90% of cases, about 43% of patients with RDD and associated adenopathy present with ≥ 1 site of extranodal involvement, and only 23% of patients with RDD present with isolated extranodal sites without adenopathy.9 As was the case with our patient, the most common extranodal sites are found in the skin and soft tissue (16%).6,9 However, histopathologic diagnosis of RDD in a lipoma is exceedingly rare. We found only 1 other case report of a patient with a history of multiple lipomas who developed a new solitary nodule that was excised and demonstrated RDD upon immunohistochemical staining.4 There has been no documented association between multiple lipomas and RDD.4

Histologically, RDD is often characterized by emperipolesis (the presence of an intact cell within the cytoplasm of anther cell) and a mixed cell infiltrate that includes S100 positive histiocytes, mononuclear cells, plasma cells, and lymphocytes.10 Despite these shared histologic features among the various phenotypes of RDD, other type-specific characteristics may also be present. When compared to nodal RDD, extranodal disease tends to demonstrate a lack of nodal architecture, more fibrosis and collagen deposition, fewer RDD cells, a lower degree of emperipolesis, and alternating pale (histiocyte rich) and dark (lymphocyte rich) regions with notable polygonal histiocytes arranged in a storiform pattern.5,10

Our patient’s histology showed an overall IgG4:IgG plasma cell ratio > 40%. RDD frequently presents with IgG4-positive plasma cells, which has confounded the diagnosis of IgG4-related diseases and hyper-IgG4 disease.11 Given this association, the Histiocyte Society revised classification recommends that all cases of RDD be evaluated for IgG4-positive cell infiltration.2,3 Further discussion on this matter was recently provided after an expert panel published a consensus statement in 2015 detailing the evaluation of IgG4. The panel advocates for stricter terminology and criteria on this issue, advises that isolated IgG4-positive plasma cells are nonspecific, and states that the diagnosis of IgG4 disease should be based on careful judgment and correlation with the clinical scenario and supportive findings.12 Therefore, while IgG4 positivity continues to be misleading in RDD cases, further evaluation for IgG4 disease is recommended. 

Sporadic RDD is usually self-limited with a reported remission rate of up to 50%, according to a case series of 80 patients with RDD.13 This leads to the recommendation of a period of watchful monitoring in patients with limited disease.13 In patients with unifocal extranodal disease, surgical excision has shown positive remission results; however, local recurrence of soft tissue lesions can occur at a rate of 21.4% to 51%.14 Although initiation of systemic therapy should be considered in patients with recurrent disease, there is currently no standardized regimen or medication of choice for treatment. Treatment with steroids, including prednisone 40 to 70 mg daily or dexamethasone 8 to 20 mg daily, have been shown to be effective in reducing the nodal size and symptoms, especially in cases of nonresectable multifocal extranodal disease of the central nervous system, bone, and orbital.7,15,16 However, cases of orbital, tracheal, renal, or soft tissue RDD have reported failure in treatment with steroids.17,18

According to the consensus recommendations for the treatment of RDD released in 2018, treatment with chemotherapy has shown mixed results. Anthracycline and alkylating agents have shown minimal efficacy, but combination regimens with vinca alkaloids, methotrexate, and 6-mercaptopurine have helped patients experience remission.19,20 Due to the rarity of RDD and lack of clinical trials, the exact efficacy of these treatment regimens remains unknown and is largely limited to case reports described within the medical literature. Treatment with nucleoside analogs, such as cladribine 2.1 to 5 mg/m2 or clofarabine 25 mg/m2 per day for 5 days every 28 days for 6 months, have shown promising results and helped achieve complete remission in patients with refractory or recurrent RDD.7,21-23 Immunomodulator therapies including TNF-α inhibitor, such as thalidomide and lenalidomide, have also shown to be effective, particularly in patients with refractory disease.24,25 Low-dose thalidomide (100 mg daily) was effective for cases of refractory cutaneous RDD, though no standard dosing regimen exists. Lenalidomide has shown to be effective in patients with multiple refractory nodal or bone RDD, but is associated with more complications given that it is more myelosuppressive than thalidomide.7 Radiotherapy has also been initiated in patients with refractory soft tissue disease or persistent symptoms after resection and in patients who are not candidates for surgery or systemic therapy, though no standard doses of radiotherapy have been established.7,26,27

Conclusions

RDD is a rare histiocytic disorder that presents in a wide range of age groups, different locations in the body, and with variable disease behavior. Multidisciplinary management of the disease and research for mutations and microenvironment of RDD is needed to better understand its clinicopathological nature and improve targeted novel therapies.

Acknowledgments

The authors thank Veterans Affairs Central Texas Health Care Section Chief of Rheumatology, Swastika Jha, MD, for her guidance in this case and Bo Wang, MD, for his preparation of the pathological specimens. 

Rosai Dorfman disease (RDD) is a rare non-Langerhans cell histiocytosis first described in 1965 by Destombes and again in 1969 by Rosai and Dorfman to depict patients who presented with massive cervical lymphadenopathy.1 The classification for histiocytosis was revised in 2016 based on new insights into the pathologic, genetic, and molecular features of RDD.2,3 Now, RDD is listed under the R group, which includes familial, sporadic, classical (nodal), extranodal RDD, and other noncutaneous, non-Langerhans cell histiocytosis.3 Cutaneous RDD is classified under the C group and typically presents as painless papules, plaques, or nodules without significant lymphadenopathy, or systemic symptoms usually seen in the presentation of RDD.4

The etiology of RDD is poorly understood, although an underlying infectious or genetic component is suspected.5 Several pathogens—including human herpesvirus 6, parvovirus B19, Epstein-Barr virus, cytomegalovirus, Brucella, and Klebsiella—have all been investigated. A link to kinase mutations has been described in nodal and extranodal RDD; however, the molecular profile of cutaneous RDD remains unknown.2 Histologic findings for RDD typically include cells that are S100 positive, CD68 positive, and CD163 positive, and CD1a and langerin (CD207) negative, thus excluding Langerhans cell histiocytosis.2 The hallmark finding of RDD is emperipolesis, which results from “histiocyte-mediated phagocytosis of intact lymphocytes and other immune cells.”6 Immunoglobulin G (Ig) G4-positive plasma cells are also common, but the significance of this finding is controversial. We present a case of a patient with recurrent RDD within a right medial thigh lipoma and include a literature review to explore the significance of histologic findings and various treatment options in the setting of emerging treatment and diagnostic criteria.

 

Case Presentation

A 56-year-old African American male was evaluated in the rheumatology clinic at the Central Texas Veterans Affairs Medical Center in Temple, Texas, in 2022 for a cutaneous mass of his right medial thigh. The patient previously reported the onset of a right medial thigh mass in 2005 after he had been deployed in Iraq for about 1 year. A biopsy of the mass from 2005 showed infiltration of plasma cells, lymphocytes, and histiocytes and occasional neutrophils with noted reactivity of S100 protein and CD163, but not CD1a. The patient’s original biopsy report from March 2005 was obtained secondhand from an addendum to a Dermatology Consult note. Surgical excision of the mass was not performed until 2012 and systemic therapy was not initiated.

figure_1.png

In 2021, the mass recurred and gradually increased in size, prompting a second surgical removal. Pathology results from the 2021 mass showed a lipoma with areas of fibrosis with a mixed inflammatory cell infiltrate, including abundant lymphocytes, plasma cells, occasional hemosiderin-laden histiocytes, and clusters of enlarged histiocytes with foamy to pale eosinophilic, finely granular cytoplasm, and large, round, vesicular nuclei with prominent nucleoli. Emperipolesis was also present (Figure 1).

figure_2.png

Special immunohistochemical staining showed most of the lymphocytes were CD20 positive B-cells with a minority of CD3 positive T-cells. Histiocytes were CD163 positive and CD68 positive with patchy reactivity for S100 protein. The plasma cells were CD138 positive. There were > 125 IgG4-positive plasma cells present in a single high-powered field and the overall IgG4:IgG plasma cell ratio was > 40%. Pertinent imaging included a whole-body positron emission tomography/computed tomography (PET/CT) hypermetabolic activity scan of a small right femoral lymph node (9 mm) and nearby medial right femoral lymph node (13 mm) (Figure 2A). A well-defined mass in the medial aspect of the right thigh (2.5 cm x 3.2 cm x 3.9 cm) and a cutaneous/subcutaneous lesion of the anterior medial aspect of the proximal right thigh superior to the mass (2.9 cm) were also evident on imaging (Figure 2B). Each area of hypermetabolic activity had decreased in size and activity when compared to a previous PET/CT obtained 1 month earlier. There was no evidence of skeletal malignancy. A physical examination did not reveal any other soft tissue masses, palpable lymphadenopathy, or areas of skin involvement. Given the patient’s reassuring imaging findings and a lack of any new physical examination findings, no systemic therapy was initiated, and following shared decision making, the patient agreed to a period of watchful waiting. 

 

 

Discussion

RDD is rare with a prevalence of 1:200,000. It has been reported that multisystem involvement occurs in 19% of cases and the prognosis of RDD correlates with the number of extranodal systems involved in the disease process.7 Although sporadic RDD is usually self-limited with favorable outcomes, it is estimated that 10% of patients may die of RDD due to direct complications, infections, and amyloidosis.2,7 RDD commonly affects young male children and young adults with a mean age of 20 years and has a higher incidence among African American children.2,7,8 Although patients with RDD present bilateral, painless cervical lymphadenopathy in 90% of cases, about 43% of patients with RDD and associated adenopathy present with ≥ 1 site of extranodal involvement, and only 23% of patients with RDD present with isolated extranodal sites without adenopathy.9 As was the case with our patient, the most common extranodal sites are found in the skin and soft tissue (16%).6,9 However, histopathologic diagnosis of RDD in a lipoma is exceedingly rare. We found only 1 other case report of a patient with a history of multiple lipomas who developed a new solitary nodule that was excised and demonstrated RDD upon immunohistochemical staining.4 There has been no documented association between multiple lipomas and RDD.4

Histologically, RDD is often characterized by emperipolesis (the presence of an intact cell within the cytoplasm of anther cell) and a mixed cell infiltrate that includes S100 positive histiocytes, mononuclear cells, plasma cells, and lymphocytes.10 Despite these shared histologic features among the various phenotypes of RDD, other type-specific characteristics may also be present. When compared to nodal RDD, extranodal disease tends to demonstrate a lack of nodal architecture, more fibrosis and collagen deposition, fewer RDD cells, a lower degree of emperipolesis, and alternating pale (histiocyte rich) and dark (lymphocyte rich) regions with notable polygonal histiocytes arranged in a storiform pattern.5,10

Our patient’s histology showed an overall IgG4:IgG plasma cell ratio > 40%. RDD frequently presents with IgG4-positive plasma cells, which has confounded the diagnosis of IgG4-related diseases and hyper-IgG4 disease.11 Given this association, the Histiocyte Society revised classification recommends that all cases of RDD be evaluated for IgG4-positive cell infiltration.2,3 Further discussion on this matter was recently provided after an expert panel published a consensus statement in 2015 detailing the evaluation of IgG4. The panel advocates for stricter terminology and criteria on this issue, advises that isolated IgG4-positive plasma cells are nonspecific, and states that the diagnosis of IgG4 disease should be based on careful judgment and correlation with the clinical scenario and supportive findings.12 Therefore, while IgG4 positivity continues to be misleading in RDD cases, further evaluation for IgG4 disease is recommended. 

Sporadic RDD is usually self-limited with a reported remission rate of up to 50%, according to a case series of 80 patients with RDD.13 This leads to the recommendation of a period of watchful monitoring in patients with limited disease.13 In patients with unifocal extranodal disease, surgical excision has shown positive remission results; however, local recurrence of soft tissue lesions can occur at a rate of 21.4% to 51%.14 Although initiation of systemic therapy should be considered in patients with recurrent disease, there is currently no standardized regimen or medication of choice for treatment. Treatment with steroids, including prednisone 40 to 70 mg daily or dexamethasone 8 to 20 mg daily, have been shown to be effective in reducing the nodal size and symptoms, especially in cases of nonresectable multifocal extranodal disease of the central nervous system, bone, and orbital.7,15,16 However, cases of orbital, tracheal, renal, or soft tissue RDD have reported failure in treatment with steroids.17,18

According to the consensus recommendations for the treatment of RDD released in 2018, treatment with chemotherapy has shown mixed results. Anthracycline and alkylating agents have shown minimal efficacy, but combination regimens with vinca alkaloids, methotrexate, and 6-mercaptopurine have helped patients experience remission.19,20 Due to the rarity of RDD and lack of clinical trials, the exact efficacy of these treatment regimens remains unknown and is largely limited to case reports described within the medical literature. Treatment with nucleoside analogs, such as cladribine 2.1 to 5 mg/m2 or clofarabine 25 mg/m2 per day for 5 days every 28 days for 6 months, have shown promising results and helped achieve complete remission in patients with refractory or recurrent RDD.7,21-23 Immunomodulator therapies including TNF-α inhibitor, such as thalidomide and lenalidomide, have also shown to be effective, particularly in patients with refractory disease.24,25 Low-dose thalidomide (100 mg daily) was effective for cases of refractory cutaneous RDD, though no standard dosing regimen exists. Lenalidomide has shown to be effective in patients with multiple refractory nodal or bone RDD, but is associated with more complications given that it is more myelosuppressive than thalidomide.7 Radiotherapy has also been initiated in patients with refractory soft tissue disease or persistent symptoms after resection and in patients who are not candidates for surgery or systemic therapy, though no standard doses of radiotherapy have been established.7,26,27

Conclusions

RDD is a rare histiocytic disorder that presents in a wide range of age groups, different locations in the body, and with variable disease behavior. Multidisciplinary management of the disease and research for mutations and microenvironment of RDD is needed to better understand its clinicopathological nature and improve targeted novel therapies.

Acknowledgments

The authors thank Veterans Affairs Central Texas Health Care Section Chief of Rheumatology, Swastika Jha, MD, for her guidance in this case and Bo Wang, MD, for his preparation of the pathological specimens. 

References

1. Goyal G, Ravindran A, Young JR, et al. Clinicopathological features, treatment approaches, and outcomes in Rosai-Dorfman disease. Haematologica. 2020;105(2):348-357. Published 2020 Jan 31. doi:10.3324/haematol.2019.219626

2. Bruce-Brand C, Schneider JW, Schubert P. Rosai-Dorfman disease: an overview. J Clin Pathol. 2020;73(11):697-705. doi:10.1136/jclinpath-2020-206733

3. Emile JF, Abla O, Fraitag S, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. 2016;127(22):2672-2681. doi:10.1182/blood-2016-01-690636

4. Farooq U, Chacon AH, Vincek V, Elgart G. Purely cutaneous rosai-dorfman disease with immunohistochemistry. Indian J Dermatol. 2013;58(6):447-450. doi:10.4103/0019-5154.119953

5. Ma H, Zheng Y, Zhu G, Wu J, Lu C, Lai W. Rosai-dorfman disease with massive cutaneous nodule on the shoulder and back. Ann Dermatol. 2015;27(1):71-75. doi:10.5021/ad.2015.27.1.71

6. Deen IU, Chittal A, Badro N, Jones R, Haas C. Extranodal Rosai-Dorfman Disease- a Review of Diagnostic Testing and Management. J Community Hosp Intern Med Perspect. 2022;12(2):18-22. Published 2022 Apr 12. doi:10.55729/2000-9666.1032

7. Oussama A, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877-2890. doi: 10.1182/blood-2018-03-839753

8. Foucar E, Rosai J, Dorfman R. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): review of the entity. Semin Diagn Pathol. 1990;7(1):19-73.

9. Gaitonde S. Multifocal, extranodal sinus histiocytosis with massive lymphadenopathy: an overview. Arch Pathol Lab Med. 2007;131(7):1111-1121. doi:10.5858/2007-131-1117-MESHWM

10. Betini N, Munger AM, Rottmann D, Haims A, Costa J, Lindskog DM. Rare presentation of Rosai-Dorfman disease in soft tissue: diagnostic findings and surgical treatment. Case Rep Surg. 2022;2022:8440836. Published 2022 Mar 30. doi:10.1155/2022/8440836

11. Menon MP, Evbuomwan MO, Rosai J, Jaffe ES, Pittaluga S. A subset of Rosai-Dorfman disease cases show increased IgG4-positive plasma cells: another red herring or a true association with IgG4-related disease? Histopathology. 2014;64(3):455-459. doi:10.1111/his.12274

12. Khosroshahi A, Wallace ZS, Crowe JL, et al. International consensus guidance statement on the management and treatment of IgG4-related disease. Arthritis Rheumatol. 2015;67(7):1688-1699. doi:10.1002/art.39132

13. Pulsoni A, Anghel G, Falcucci P, et al. Treatment of sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): report of a case and literature review. Am J Hematol. 2002;69(1):67-71. doi:10.1002/ajh.10008

14. Montgomery EA, Meis JM, Firzzera G. Rosai-Dorfman disease of soft tissue. Am J Surg Pathol. 1992;16(2):122-129. doi:10.1097/00000478-199202000-00004

15. Z’Graggen WJ, Sturzenegger M, Mariani L, Keserue B, Kappeler A, Vajtai I. Isolated Rosai-Dorfman disease of intracranial meninges. Pathol Res Pract. 2006;202(3):165-170. doi:10.1016/j.prp.2005.11.004

16. Shulman S, Katzenstein H, Abramowsky C, Broecker J, Wulkan M, Shehata B. Unusual presentation of Rosai-Dorfman disease (RDD) in the bone in adolescents. Fetal Pediatr Pathol. 2011;30(6):442-447. doi:10.3109/15513815.2011.61887317. Ottaviano G, Doro D, Marioni G, et al. Extranodal Rosai-Dorfman disease: involvement of eye, nose and trachea. Acta Otolaryngol. 2006;126(6):657-660. doi:10.1080/00016480500452582

18. Sakallioglu O, Gok F, Kalman S, et al. Minimal change nephropathy in a 7-year-old boy with Rosai-Dorfman disease. J Nephrol. 2006;19(2):211-214.

19. Jabali Y, Smrcka V, Pradna J. Rosai-Dorfman disease: successful long-term results by combination chemotherapy with prednisone, 6-mercaptopurine, methotrexate, and vinblastine: a case report. Int J Surg Pathol. 2005;13(3):285-289. doi:10.1177/106689690501300311

20. Abla O, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877-2890. doi:10.1182/blood-2018-03-839753

21. Konca C, Özkurt ZN, Deger M, Akı Z, Yagcı M. Extranodal multifocal Rosai-Dorfman disease: response to 2-chlorodeoxyadenosine treatment. Int J Hematol. 2009;89(1):58-62. doi:10.1007/s12185-008-0192-2

22. Aouba A, Terrier B, Vasiliu V, et al. Dramatic clinical efficacy of cladribine in Rosai-Dorfman disease and evolution of the cytokine profile: towards a new therapeutic approach. Haematologica. 2006;91(12 Suppl):ECR52.

23. Tasso M, Esquembre C, Blanco E, Moscardó C, Niveiro M, Payá A. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) treated with 2-chlorodeoxyadenosine. Pediatr Blood Cancer. 2006;47(5):612-615. doi:10.1002/pbc.20668

24. Chen E, Pavlidakey P, Sami N. Rosai-Dorfman disease successfully treated with thalidomide. JAAD Case Reports. 2016;2(5):369-372. Published 2016 Sep 28. doi:10.1016/j.jdcr.2016.08.006

25. Rubinstein M, Assal A, Scherba M, et al. Lenalidomide in the treatment of Rosai Dorfman disease-a first in use report. Am J Hematol. 2016;91(2):E1. doi:10.1002/ajh.24225

26. Sandoval-Sus JD, Sandoval-Leon AC, Chapman JR, et al. Rosai-Dorfman disease of the central nervous system: report of 6 cases and review of the literature. Medicine (Baltimore). 2014;93(3):165-175. doi:10.1097/MD.0000000000000030

27. Paryani NN, Daugherty LC, O’Connor MI, Jiang L. Extranodal Rosai-Dorfman disease of the bone treated with surgery and radiotherapy. Rare Tumors. 2014;6(4):5531. Published 2014 Dec 11. doi:10.4081/rt.2014.5531

References

1. Goyal G, Ravindran A, Young JR, et al. Clinicopathological features, treatment approaches, and outcomes in Rosai-Dorfman disease. Haematologica. 2020;105(2):348-357. Published 2020 Jan 31. doi:10.3324/haematol.2019.219626

2. Bruce-Brand C, Schneider JW, Schubert P. Rosai-Dorfman disease: an overview. J Clin Pathol. 2020;73(11):697-705. doi:10.1136/jclinpath-2020-206733

3. Emile JF, Abla O, Fraitag S, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. 2016;127(22):2672-2681. doi:10.1182/blood-2016-01-690636

4. Farooq U, Chacon AH, Vincek V, Elgart G. Purely cutaneous rosai-dorfman disease with immunohistochemistry. Indian J Dermatol. 2013;58(6):447-450. doi:10.4103/0019-5154.119953

5. Ma H, Zheng Y, Zhu G, Wu J, Lu C, Lai W. Rosai-dorfman disease with massive cutaneous nodule on the shoulder and back. Ann Dermatol. 2015;27(1):71-75. doi:10.5021/ad.2015.27.1.71

6. Deen IU, Chittal A, Badro N, Jones R, Haas C. Extranodal Rosai-Dorfman Disease- a Review of Diagnostic Testing and Management. J Community Hosp Intern Med Perspect. 2022;12(2):18-22. Published 2022 Apr 12. doi:10.55729/2000-9666.1032

7. Oussama A, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877-2890. doi: 10.1182/blood-2018-03-839753

8. Foucar E, Rosai J, Dorfman R. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): review of the entity. Semin Diagn Pathol. 1990;7(1):19-73.

9. Gaitonde S. Multifocal, extranodal sinus histiocytosis with massive lymphadenopathy: an overview. Arch Pathol Lab Med. 2007;131(7):1111-1121. doi:10.5858/2007-131-1117-MESHWM

10. Betini N, Munger AM, Rottmann D, Haims A, Costa J, Lindskog DM. Rare presentation of Rosai-Dorfman disease in soft tissue: diagnostic findings and surgical treatment. Case Rep Surg. 2022;2022:8440836. Published 2022 Mar 30. doi:10.1155/2022/8440836

11. Menon MP, Evbuomwan MO, Rosai J, Jaffe ES, Pittaluga S. A subset of Rosai-Dorfman disease cases show increased IgG4-positive plasma cells: another red herring or a true association with IgG4-related disease? Histopathology. 2014;64(3):455-459. doi:10.1111/his.12274

12. Khosroshahi A, Wallace ZS, Crowe JL, et al. International consensus guidance statement on the management and treatment of IgG4-related disease. Arthritis Rheumatol. 2015;67(7):1688-1699. doi:10.1002/art.39132

13. Pulsoni A, Anghel G, Falcucci P, et al. Treatment of sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): report of a case and literature review. Am J Hematol. 2002;69(1):67-71. doi:10.1002/ajh.10008

14. Montgomery EA, Meis JM, Firzzera G. Rosai-Dorfman disease of soft tissue. Am J Surg Pathol. 1992;16(2):122-129. doi:10.1097/00000478-199202000-00004

15. Z’Graggen WJ, Sturzenegger M, Mariani L, Keserue B, Kappeler A, Vajtai I. Isolated Rosai-Dorfman disease of intracranial meninges. Pathol Res Pract. 2006;202(3):165-170. doi:10.1016/j.prp.2005.11.004

16. Shulman S, Katzenstein H, Abramowsky C, Broecker J, Wulkan M, Shehata B. Unusual presentation of Rosai-Dorfman disease (RDD) in the bone in adolescents. Fetal Pediatr Pathol. 2011;30(6):442-447. doi:10.3109/15513815.2011.61887317. Ottaviano G, Doro D, Marioni G, et al. Extranodal Rosai-Dorfman disease: involvement of eye, nose and trachea. Acta Otolaryngol. 2006;126(6):657-660. doi:10.1080/00016480500452582

18. Sakallioglu O, Gok F, Kalman S, et al. Minimal change nephropathy in a 7-year-old boy with Rosai-Dorfman disease. J Nephrol. 2006;19(2):211-214.

19. Jabali Y, Smrcka V, Pradna J. Rosai-Dorfman disease: successful long-term results by combination chemotherapy with prednisone, 6-mercaptopurine, methotrexate, and vinblastine: a case report. Int J Surg Pathol. 2005;13(3):285-289. doi:10.1177/106689690501300311

20. Abla O, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877-2890. doi:10.1182/blood-2018-03-839753

21. Konca C, Özkurt ZN, Deger M, Akı Z, Yagcı M. Extranodal multifocal Rosai-Dorfman disease: response to 2-chlorodeoxyadenosine treatment. Int J Hematol. 2009;89(1):58-62. doi:10.1007/s12185-008-0192-2

22. Aouba A, Terrier B, Vasiliu V, et al. Dramatic clinical efficacy of cladribine in Rosai-Dorfman disease and evolution of the cytokine profile: towards a new therapeutic approach. Haematologica. 2006;91(12 Suppl):ECR52.

23. Tasso M, Esquembre C, Blanco E, Moscardó C, Niveiro M, Payá A. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) treated with 2-chlorodeoxyadenosine. Pediatr Blood Cancer. 2006;47(5):612-615. doi:10.1002/pbc.20668

24. Chen E, Pavlidakey P, Sami N. Rosai-Dorfman disease successfully treated with thalidomide. JAAD Case Reports. 2016;2(5):369-372. Published 2016 Sep 28. doi:10.1016/j.jdcr.2016.08.006

25. Rubinstein M, Assal A, Scherba M, et al. Lenalidomide in the treatment of Rosai Dorfman disease-a first in use report. Am J Hematol. 2016;91(2):E1. doi:10.1002/ajh.24225

26. Sandoval-Sus JD, Sandoval-Leon AC, Chapman JR, et al. Rosai-Dorfman disease of the central nervous system: report of 6 cases and review of the literature. Medicine (Baltimore). 2014;93(3):165-175. doi:10.1097/MD.0000000000000030

27. Paryani NN, Daugherty LC, O’Connor MI, Jiang L. Extranodal Rosai-Dorfman disease of the bone treated with surgery and radiotherapy. Rare Tumors. 2014;6(4):5531. Published 2014 Dec 11. doi:10.4081/rt.2014.5531

Issue
Federal Practitioner - 41(5)s
Issue
Federal Practitioner - 41(5)s
Page Number
S20
Page Number
S20
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>0524 AVAHO RDD</fileName> <TBEID>0C02F4C6.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F4C6</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240428T220046</firstPublished> <LastPublished>20240428T220046</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240428T220046</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>John Corbyn Cravero, MDa; Samar Ibrahim, 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>Rosai Dorfman disease (RDD) is a rare non-Langerhans cell histiocytosis first described in 1965 by Destombes and again in 1969 by Rosai and Dorfman to depict pa</metaDescription> <articlePDF/> <teaserImage/> <title>Recurrent Soft Tissue Rosai Dorfman Disease of Right Medial Thigh Lipoma With Lymph Node Involvement</title> <deck/> <eyebrow>Case In Point</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2953</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case in Point | 2953<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">45</term> </sections> <topics> <term>27442</term> <term canonical="true">263</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Recurrent Soft Tissue Rosai Dorfman Disease of Right Medial Thigh Lipoma With Lymph Node Involvement</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Rosai Dorfman disease is a rare non-Langerhans cell histiocytosis that affects lymph nodes, soft tissues, and other organs. The etiology of Rosai Dorfman disease is poorly understood, though it may involve an immunologic processes or infection. Treatment varies according to the disease presentation.<br/><br/><b>Case Presentation:</b> A male aged 56 years was evaluated for a cutaneous mass on his right medial thigh. Initially, the patient received surgical debulking with subsequent observation and no systemic therapy. However, the mass recurred, prompting another surgical removal 9 years after the initial surgery. A mass biopsy showed infiltration of plasma cells, lymphocytes, histiocytes, and occasional neutrophils with noted reactivity of S-100 protein and CD163, but not CD1a. No systemic therapy was initiated, and the patient agreed to a period of watchful waiting.<br/><br/><b>Conclusions:</b> Rosai Dorfman disease of soft tissue occurs in older adults and is often associated with soft tissue abnormalities, and more rarely, in lipomas. Multidisciplinary management of the disease and research for mutations and microenvironment is needed to better understand its clinicopathological nature and improve novel therapies.</p> <p>Rosai Dorfman disease (RDD) is a rare non-Langerhans cell histiocytosis first described in 1965 by Destombes and again in 1969 by Rosai and Dorfman to depict patients who presented with massive cervical lymphadenopathy.<sup>1</sup> The classification for histiocytosis was revised in 2016 based on new insights into the pathologic, genetic, and molecular features of RDD.<sup>2,3</sup> Now, RDD is listed under the R group, which includes familial, sporadic, classical (nodal), extranodal RDD, and other noncutaneous, non-Langerhans cell histiocytosis.<sup>3</sup> Cutaneous RDD is classified under the C group and typically presents as painless papules, plaques, or nodules without significant lymphadenopathy, or systemic symptoms usually seen in the presentation of RDD.<sup>4</sup></p> <p>The etiology of RDD is poorly understood, although an underlying infectious or genetic component is suspected.<sup>5</sup> Several pathogens—including human herpesvirus 6, parvovirus B19, Epstein-Barr virus, cytomegalovirus, <i>Brucella</i>, and <i>Klebsiella</i>—have all been investigated. A link to kinase mutations has been described in nodal and extranodal RDD; however, the molecular profile of cutaneous RDD remains unknown.<sup>2</sup> Histologic findings for RDD typically include cells that are S100 positive, CD68 positive, and CD163 positive, and CD1a and langerin (CD207) negative, thus excluding Langerhans cell histiocytosis.<sup>2</sup> The hallmark finding of RDD is emperipolesis, which results from “histiocyte-mediated phagocytosis of intact lymphocytes and other immune cells.”<sup>6</sup> Immunoglobulin G (Ig) G4-positive plasma cells are also common, but the significance of this finding is controversial. We present a case of a patient with recurrent RDD within a right medial thigh lipoma and include a literature review to explore the significance of histologic findings and various treatment options in the setting of emerging treatment and diagnostic criteria.</p> <h2>Case PRESENTATION</h2> <p>A 56-year-old African American male was evaluated in the rheumatology clinic at the Central Texas Veterans Affairs Medical Center in Temple, Texas, in 2022 for a cutaneous mass of his right medial thigh. The patient previously reported the onset of a right medial thigh mass in 2005 after he had been deployed in Iraq for about 1 year. A biopsy of the mass from 2005 showed infiltration of plasma cells, lymphocytes, and histiocytes and occasional neutrophils with noted reactivity of S100 protein and CD163, but not CD1a. The patient’s original biopsy report from March 2005 was obtained secondhand from an addendum to a Dermatology Consult note. Surgical excision of the mass was not performed until 2012 and systemic therapy was not initiated.</p> <p>In 2021, the mass recurred and gradually increased in size, prompting a second surgical removal. Pathology results from the 2021 mass showed a lipoma with areas of fibrosis with a mixed inflammatory cell infiltrate, including abundant lymphocytes, plasma cells, occasional hemosiderin-laden histiocytes, and clusters of enlarged histiocytes with foamy to pale eosinophilic, finely granular cytoplasm, and large, round, vesicular nuclei with prominent nucleoli. Emperipolesis was also present (Figure 1).<br/><br/>Special immunohistochemical staining showed most of the lymphocytes were CD20 positive B-cells with a minority of CD3 positive T-cells. Histiocytes were CD163 positive and CD68 positive with patchy reactivity for S100 protein. The plasma cells were CD138 positive. There were &gt; 125 IgG4-positive plasma cells present in a single high-powered field and the overall IgG4:IgG plasma cell ratio was &gt; 40%. Pertinent imaging included a whole-body positron emission tomography/computed tomography (PET/CT) hypermetabolic activity scan of a small right femoral lymph node (9 mm) and nearby medial right femoral lymph node (13 mm) (Figure 2A). A well-defined mass in the medial aspect of the right thigh (2.5 cm x 3.2 cm x 3.9 cm) and a cutaneous/subcutaneous lesion of the anterior medial aspect of the proximal right thigh superior to the mass (2.9 cm) were also evident on imaging (Figure 2B). Each area of hypermetabolic activity had decreased in size and activity when compared to a previous PET/CT obtained 1 month earlier. There was no evidence of skeletal malignancy. A physical examination did not reveal any other soft tissue masses, palpable lymphadenopathy, or areas of skin involvement. Given the patient’s reassuring imaging findings and a lack of any new physical examination findings, no systemic therapy was initiated, and following shared decision making, the patient agreed to a period of watchful waiting. </p> <h2>Discussion</h2> <p>RDD is rare with a prevalence of 1:200,000. It has been reported that multisystem involvement occurs in 19% of cases and the prognosis of RDD correlates with the number of extranodal systems involved in the disease process.<sup>7</sup> Although sporadic RDD is usually self-limited with favorable outcomes, it is estimated that 10% of patients may die of RDD due to direct complications, infections, and amyloidosis.<sup>2,7</sup> RDD commonly affects young male children and young adults with a mean age of 20 years and has a higher incidence among African American children.<sup>2,7,8</sup> Although patients with RDD present bilateral, painless cervical lymphadenopathy in 90% of cases, about 43% of patients with RDD and associated adenopathy present with ≥ 1 site of extranodal involvement, and only 23% of patients with RDD present with isolated extranodal sites without adenopathy.<sup>9</sup> As was the case with our patient, the most common extranodal sites are found in the skin and soft tissue (16%).<sup>6,9</sup> However, histopathologic diagnosis of RDD in a lipoma is exceedingly rare. We found only 1 other case report of a patient with a history of multiple lipomas who developed a new solitary nodule that was excised and demonstrated RDD upon immunohistochemical staining.<sup>4</sup> There has been no documented association between multiple lipomas and RDD.<sup>4</sup></p> <p>Histologically, RDD is often characterized by emperipolesis (the presence of an intact cell within the cytoplasm of anther cell) and a mixed cell infiltrate that includes S100 positive histiocytes, mononuclear cells, plasma cells, and lymphocytes.<sup>10</sup> Despite these shared histologic features among the various phenotypes of RDD, other type-specific characteristics may also be present. When compared to nodal RDD, extranodal disease tends to demonstrate a lack of nodal architecture, more fibrosis and collagen deposition, fewer RDD cells, a lower degree of emperipolesis, and alternating pale (histiocyte rich) and dark (lymphocyte rich) regions with notable polygonal histiocytes arranged in a storiform pattern.<sup>5,10</sup> <br/><br/>Our patient’s histology showed an overall IgG4:IgG plasma cell ratio &gt; 40%. RDD frequently presents with IgG4-positive plasma cells, which has confounded the diagnosis of IgG4-related diseases and hyper-IgG4 disease.<sup>11</sup> Given this association, the Histiocyte Society revised classification recommends that all cases of RDD be evaluated for IgG4-positive cell infiltration.<sup>2,3</sup> Further discussion on this matter was recently provided after an expert panel published a consensus statement in 2015 detailing the evaluation of IgG4. The panel advocates for stricter terminology and criteria on this issue, advises that isolated IgG4-positive plasma cells are nonspecific, and states that the diagnosis of IgG4 disease should be based on careful judgment and correlation with the clinical scenario and supportive findings.<sup>12</sup> Therefore, while IgG4 positivity continues to be misleading in RDD cases, further evaluation for IgG4 disease is recommended. <br/><br/>Sporadic RDD is usually self-limited with a reported remission rate of up to 50%, according to a case series of 80 patients with RDD.<sup>13</sup> This leads to the recommendation of a period of watchful monitoring in patients with limited disease.<sup>13</sup> In patients with unifocal extranodal disease, surgical excision has shown positive remission results; however, local recurrence of soft tissue lesions can occur at a rate of 21.4% to 51%.<sup>14</sup> Although initiation of systemic therapy should be considered in patients with recurrent disease, there is currently no standardized regimen or medication of choice for treatment. Treatment with steroids, including prednisone 40 to 70 mg daily or dexamethasone 8 to 20 mg daily, have been shown to be effective in reducing the nodal size and symptoms, especially in cases of nonresectable multifocal extranodal disease of the central nervous system, bone, and orbital.<sup>7,15,16</sup> However, cases of orbital, tracheal, renal, or soft tissue RDD have reported failure in treatment with steroids.<sup>17,18</sup>According to the consensus recommendations for the treatment of RDD released in 2018, treatment with chemotherapy has shown mixed results. Anthracycline and alkylating agents have shown minimal efficacy, but combination regimens with vinca alkaloids, methotrexate, and 6-mercaptopurine have helped patients experience remission.<sup>19,20</sup> Due to the rarity of RDD and lack of clinical trials, the exact efficacy of these treatment regimens remains unknown and is largely limited to case reports described within the medical literature. Treatment with nucleoside analogs, such as cladribine 2.1 to 5 mg/m<sup>2</sup> or clofarabine 25 mg/m<sup>2</sup> per day for 5 days every 28 days for 6 months, have shown promising results and helped achieve complete remission in patients with refractory or recurrent RDD.<sup>7,21-23</sup> Immunomodulator therapies including TNF-α inhibitor, such as thalidomide and lenalidomide, have also shown to be effective, particularly in patients with refractory disease.<sup>24,25</sup> Low-dose thalidomide (100 mg daily) was effective for cases of refractory cutaneous RDD, though no standard dosing regimen exists. Lenalidomide has shown to be effective in patients with multiple refractory nodal or bone RDD, but is associated with more complications given that it is more myelosuppressive than thalidomide.<sup>7</sup> Radiotherapy has also been initiated in patients with refractory soft tissue disease or persistent symptoms after resection and in patients who are not candidates for surgery or systemic therapy, though no standard doses of radiotherapy have been established.<sup>7,26,27</sup></p> <h2>ConclusionS</h2> <p>RDD is a rare histiocytic disorder that presents in a wide range of age groups, different locations in the body, and with variable disease behavior. Multidisciplinary management of the disease and research for mutations and microenvironment of RDD is needed to better understand its clinicopathological nature and improve targeted novel therapies.</p> <p class="isub">Acknowledgments</p> <p> <em>The authors thank Veterans Affairs Central Texas Health Care Section Chief of Rheumatology, Swastika Jha, MD, for her guidance in this case and Bo Wang, MD, for his preparation of the pathological specimens. </em> </p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Baylor Scott &amp; White Medical Center – Temple, Texas</em> </p> <p class="isub">Author disclosures</p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>FederalPractitioner, </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> <p class="isub">Ethics and consent</p> <p> <em>Written informed consent for publication was obtained by the patient who was involved in this case report.</em> </p> <p class="isub">References</p> <p class="reference"> 1. Goyal G, Ravindran A, Young JR, et al. Clinicopathological features, treatment approaches, and outcomes in Rosai-Dorfman disease. <i>Haematologica</i>. 2020;105(2):348-357. Published 2020 Jan 31. doi:10.3324/haematol.2019.219626<br/><br/> 2. Bruce-Brand C, Schneider JW, Schubert P. Rosai-Dorfman disease: an overview. <i>J Clin Pathol</i>. 2020;73(11):697-705. doi:10.1136/jclinpath-2020-206733<br/><br/> 3. Emile JF, Abla O, Fraitag S, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. <i>Blood</i>. 2016;127(22):2672-2681. doi:10.1182/blood-2016-01-690636<br/><br/> 4. Farooq U, Chacon AH, Vincek V, Elgart G. Purely cutaneous rosai-dorfman disease with immunohistochemistry. <i>Indian J Dermatol</i>. 2013;58(6):447-450. doi:10.4103/0019-5154.119953<br/><br/> 5. Ma H, Zheng Y, Zhu G, Wu J, Lu C, Lai W. Rosai-dorfman disease with massive cutaneous nodule on the shoulder and back. <i>Ann Dermatol</i>. 2015;27(1):71-75. doi:10.5021/ad.2015.27.1.71<br/><br/> 6. Deen IU, Chittal A, Badro N, Jones R, Haas C. Extranodal Rosai-Dorfman Disease- a Review of Diagnostic Testing and Management. <i>J Community Hosp Intern Med Perspect</i>. 2022;12(2):18-22. Published 2022 Apr 12. doi:10.55729/2000-9666.1032<br/><br/> 7. Oussama A, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. <i>Blood</i>. 2018;131(26):2877-2890. doi: 10.1182/blood-2018-03-839753<br/><br/> 8. Foucar E, Rosai J, Dorfman R. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): review of the entity. <i>Semin Diagn Pathol</i>. 1990;7(1):19-73.<br/><br/> 9. Gaitonde S. Multifocal, extranodal sinus histiocytosis with massive lymphadenopathy: an overview. <i>Arch Pathol Lab Med</i>. 2007;131(7):1111-1121. doi:10.5858/2007-131-1117-MESHWM<br/><br/>10. Betini N, Munger AM, Rottmann D, Haims A, Costa J, Lindskog DM. Rare presentation of Rosai-Dorfman disease in soft tissue: diagnostic findings and surgical treatment. <i>Case Rep Surg</i>. 2022;2022:8440836. Published 2022 Mar 30. doi:10.1155/2022/8440836</p> <p class="reference">11. Menon MP, Evbuomwan MO, Rosai J, Jaffe ES, Pittaluga S. A subset of Rosai-Dorfman disease cases show increased IgG4-positive plasma cells: another red herring or a true association with IgG4-related disease? <i>Histopathology</i>. 2014;64(3):455-459. doi:10.1111/his.12274<br/><br/>12. Khosroshahi A, Wallace ZS, Crowe JL, et al. International consensus guidance statement on the management and treatment of IgG4-related disease. <i>Arthritis Rheumatol</i>. 2015;67(7):1688-1699. doi:10.1002/art.39132<br/><br/>13. Pulsoni A, Anghel G, Falcucci P, et al. Treatment of sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease): report of a case and literature review. <i>Am J Hematol</i>. 2002;69(1):67-71. doi:10.1002/ajh.10008<br/><br/>14. Montgomery EA, Meis JM, Firzzera G. Rosai-Dorfman disease of soft tissue. <i>Am J Surg Pathol</i>. 1992;16(2):122-129. doi:10.1097/00000478-199202000-00004<br/><br/>15. Z’Graggen WJ, Sturzenegger M, Mariani L, Keserue B, Kappeler A, Vajtai I. Isolated Rosai-Dorfman disease of intracranial meninges. <i>Pathol Res Pract</i>. 2006;202(3):165-170. doi:10.1016/j.prp.2005.11.004<br/><br/>16. Shulman S, Katzenstein H, Abramowsky C, Broecker J, Wulkan M, Shehata B. Unusual presentation of Rosai-Dorfman disease (RDD) in the bone in adolescents. <i>Fetal Pediatr Pathol</i>. 2011;30(6):442-447. doi:10.3109/15513815.2011.61887317. Ottaviano G, Doro D, Marioni G, et al. Extranodal Rosai-Dorfman disease: involvement of eye, nose and trachea. <i>Acta Otolaryngol</i>. 2006;126(6):657-660. doi:10.1080/00016480500452582<br/><br/>18. Sakallioglu O, Gok F, Kalman S, et al. Minimal change nephropathy in a 7-year-old boy with Rosai-Dorfman disease. <i>J Nephrol</i>. 2006;19(2):211-214.<br/><br/>19. Jabali Y, Smrcka V, Pradna J. Rosai-Dorfman disease: successful long-term results by combination chemotherapy with prednisone, 6-mercaptopurine, methotrexate, and vinblastine: a case report. <i>Int J Surg Pathol</i>. 2005;13(3):285-289. doi:10.1177/106689690501300311<br/><br/>20. Abla O, Jacobsen E, Picarsic J, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. <i>Blood</i>. 2018;131(26):2877-2890. doi:10.1182/blood-2018-03-839753<br/><br/>21. Konca C, Özkurt ZN, Deger M, Akı Z, Yagcı M. Extranodal multifocal Rosai-Dorfman disease: response to 2-chlorodeoxyadenosine treatment. <i>Int J Hematol</i>. 2009;89(1):58-62. doi:10.1007/s12185-008-0192-2<br/><br/>22. Aouba A, Terrier B, Vasiliu V, et al. Dramatic clinical efficacy of cladribine in Rosai-Dorfman disease and evolution of the cytokine profile: towards a new therapeutic approach. <i>Haematologica</i>. 2006;91(12 Suppl):ECR52.<br/><br/>23. Tasso M, Esquembre C, Blanco E, Moscardó C, Niveiro M, Payá A. Sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) treated with 2-chlorodeoxyadenosine. <i>Pediatr Blood Cancer</i>. 2006;47(5):612-615. doi:10.1002/pbc.20668<br/><br/>24. Chen E, Pavlidakey P, Sami N. Rosai-Dorfman disease successfully treated with thalidomide. <i>JAAD Case Reports</i>. 2016;2(5):369-372. Published 2016 Sep 28. doi:10.1016/j.jdcr.2016.08.006<br/><br/>25. Rubinstein M, Assal A, Scherba M, et al. Lenalidomide in the treatment of Rosai Dorfman disease-a first in use report. <i>Am J Hematol</i>. 2016;91(2):E1. doi:10.1002/ajh.24225<br/><br/>26. Sandoval-Sus JD, Sandoval-Leon AC, Chapman JR, et al. Rosai-Dorfman disease of the central nervous system: report of 6 cases and review of the literature. <i>Medicine (Baltimore)</i>. 2014;93(3):165-175. doi:10.1097/MD.0000000000000030<br/><br/>27. Paryani NN, Daugherty LC, O’Connor MI, Jiang L. Extranodal Rosai-Dorfman disease of the bone treated with surgery and radiotherapy. <i>Rare Tumors</i>. 2014;6(4):5531. Published 2014 Dec 11. doi:10.4081/rt.2014.5531</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
Image
Disable zoom
Off

EBER-Negative, Double-Hit High-Grade B-Cell Lymphoma Responding to Methotrexate Discontinuation

Article Type
Changed
Mon, 04/29/2024 - 19:24

High-grade B-cell lymphomas (HGBCLs) are aggressive lymphoproliferative disorders (LPDs) that require fluorescence in-situ hybridization to identify gene rearrangements within MYC and BCL2 and/or BCL6 oncogenes. Traditionally referred to as double-hit or triple-hit lymphomas, HGBCL is a newer entity in the 2016 updated World Health Organization classification of lymphoid neoplasms.1 More than 90% of patients with HGBCL present with advanced clinical features, such as central nervous system involvement, leukocytosis, or lactose dehydrogenase (LDH) greater than 3 times the upper limit of normal. Treatment outcomes with aggressive multiagent chemotherapy combined with anti-CD20–targeted therapy are generally worse for patients with double-hit disease, especially among frail patients with advanced age. Patients with underlying autoimmune and rheumatologic conditions, such as rheumatoid arthritis (RA), are at higher risk for developing LPDs. These include highly aggressive subtypes of non-Hodgkin lymphoma, such as HGBCL, likely due to cascading events secondary to chronic inflammation and/or immunosuppressive medications. These immunodeficiency-associated LPDs often express positivity for Epstein-Barr virus-encoded small RNA (EBER).

We present a case of double-hit HGBCL that was EBER negative with MYC and BCL6 rearrangements in an older veteran with RA managed with methotrexate. An excellent sustained response was observed for the patient’s stage IV double-hit HGBCL disease within 4 weeks of methotrexate discontinuation. To our knowledge, this is the first reported response to methotrexate discontinuation for a patient with HGBCL.

CASE PRESENTATION

A male veteran aged 81 years presented to the Raymond G. Murphy Veterans Affairs Medical Center (RGMVAMC) in Albuquerque, New Mexico, with an unintentional 25-pound weight loss over 18 months. Pertinent history included RA managed with methotrexate 15 mg weekly for 6 years and a previous remote seizure. The patients prior prostate cancer was treated with radiation at the time of diagnosis and ongoing androgen deprivation therapy. Initial workup with chest X-ray and chest computed tomography (CT) indicated loculated left pleural fluid collection with a suspected splenic tumor.

figure_1.png

A positron-emission tomography (PET)/CT was ordered given his history of prostate cancer, which showed potential splenic and sternal metastases with corresponding fludeoxyglucose F18 uptake (Figure 1A). Biopsy was not pursued due to the potential for splenic hemorrhage. Based on the patient’s RA and methotrexate use, the collection of findings was initially thought to represent a non-Hodgkin lymphoma, with knowledge that metastatic prostate cancer refractory to androgen deprivation therapy was possible. Because he was unable to undergo a splenic biopsy, an observation strategy involving repeat PET/CT every 6 months was started.

The surveillance PET/CT 6 months later conveyed worsened disease burden with increased avidity in the manubrium (Figure 1B). The patient’s case was discussed at the RGMVAMC tumor board, and the recommendation was to continue with surveillance follow-up imaging because image-guided biopsy might not definitively yield a diagnosis. Repeat PET/CT3 months later indicated continued worsening of disease (Figure 1C) with a rapidly enlarging hypermetabolic mass in the manubrium that extended anteriorly into the subcutaneous tissues and encased the bilateral anterior jugular veins. On physical examination, this sternal mass had become painful and was clearly evident. Additionally, increased avidity in multiple upper abdominal and retroperitoneal lymph nodes was observed.

figure_2.png

Interventional radiology was consulted to assist with a percutaneous fine-needle aspiration of the manubrial mass, which revealed a dense aggregate of large, atypical lymphocytes confirmed to be of B-cell origin (CD20 and PAX5 positive) (Figure 2). The atypical B cells demonstrated co-expression of BCL6, BCL2, MUM1, and MYC but were negative for CD30 and EBER by in situ hybridization. The overall morphologic and immunophenotypic findings were consistent with a large B-cell lymphoma. Fluorescent in-situ hybridization identified the presence of MYC and BCL6 gene rearrangements, and the mass was consequently best classified as a double-hit HGBCL.

Given the patient’s history of long-term methotrexate use, we thought the HGBCL may have reflected an immunodeficiency-associated LPD, although the immunophenotype was not classic because of the CD30 and EBER negativity. With the known toxicity and poor treatment outcomes of aggressive multiagent chemotherapy for patients with double-hit HGBCL—particularly in the older adult population—methotrexate was discontinued on a trial basis.

A PET/CT was completed 4 weeks after methotrexate was discontinued due to concerns about managing an HGBCL without chemotherapy or anti-CD20–directed therapy. The updated PET/CT showed significant improvement with marked reduction in avidity of his manubrial lesion (Figure 1D). Three months after methotrexate discontinuation, the patient remained in partial remission for his double-hit HGBCL, as evidenced by no findings of sternal mass on repeat examinations with continued decrease in hypermetabolic findings on PET/CT. The patient's RA symptoms rebounded, and rheumatology colleagues prescribed sulfasalazine and periodic steroid tapers to help control his inflammatory arthritis. Fourteen months after discontinuation of methotrexate, the patient died after developing pneumonia, which led to multisystemic organ failure.

 

 

DISCUSSION

HGBCL with MYC and BCL2 and/or BCL6 rearrangements is an aggressive LPD.1 A definitive diagnosis requires collection of morphologic and immunophenotypic evaluations of suspicious tissue. Approximately 60% of patients with HGBCL have translocations in MYC and BCL2, 20% have MYC and BCL6 translocations, and the remaining 20% have MYC, BCL2 and BCL6 translocations (triple-hit disease).1

The MYC and BCL gene rearrangements are thought to synergistically drive tumorigenesis, leading to accelerated lymphoma progression and a lesser response to standard multiagent chemotherapy than seen in diffuse large B-cell lymphoma.1-3 Consequently, there have been several attempts to increase treatment efficacy with intense chemotherapy regimens, namely DA-EPOCH-R (dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab), or by adding targeted agents, such as ibrutinib and venetoclax to a standard R-CHOP (rituximab with reduced cyclophosphamide, doxorubicin, vincristine, and prednisone) backbone.4-7 Though the standard choice of therapy for fit patients harboring HGBCL remains controversial, these aggressive regimens at standard doses are typically difficult to tolerate for patients aged > 80 years.

table.png

Patients with immunosuppression are at higher risk for developing LPDs, including aggressive B-cell non-Hodgkin lymphomas such as diffuse large B-cell lymphoma. These patients are frequently classified into 2 groups: those with underlying autoimmune conditions (RA-associated LPDs), or those who have undergone solid-organ or allogeneic hematopoietic stem-cell transplants, which drives the development of posttransplant LPDs (Table).8-11 Both types of LPDs are often EBER positive, indicating some association with Epstein-Barr virus infection driven by ongoing immunosuppression, with knowledge that this finding is not absolute and is less frequent among patients with autoimmune conditions than those with posttransplant LPD.8,12

For indolent and early-stage aggressive LPDs, reduction of immunosuppression is a reasonable frontline treatment. In fact, Tokuyama and colleagues reported a previous case in which an methotrexate-associated EBER-positive early-stage diffuse large B-cell lymphoma responded well to methotrexate withdrawal.13 For advanced, aggressive LPDs associated with immunosuppression, a combination strategy of reducing immunosuppression and initiating a standard multiagent systemic therapy such as with R-CHOP is more common. Reducing immunosuppression without adding systemic anticancer therapy can certainly be considered in patients with EBER-negative LPDs; however, there is less evidence supporting this approach in the literature.

A case series of patients with EBER-positive double-hit HGBCL has been described previously, and response rates were low despite aggressive treatment.14 The current case differs from that case series in 2 ways. First, our patient did not have EBER-positive disease despite having an HGBCL associated with RA and methotrexate use. Second, our patient had a very rapid and excellent partial response simply with methotrexate discontinuation. Aggressive treatment was considered initially; however, given the patient’s age and performance status, reduction of immunosuppression alone was considered the frontline approach.

This case indicates that methotrexate withdrawal may lead to remission in patients with double-hit lymphoma, even without clear signs of Epstein-Barr virus infection being present. We are not sure why our patient with EBER-negative HGBCL responded differently to methotrexate withdrawal than the patients in the aforementioned case series with EBER-positive disease; nevertheless, a short trial of methotrexate withdrawal with repeat imaging 4 to 8 weeks after discontinuation seems reasonable for patients who are older, frail, and seemingly not fit for more aggressive treatment.

CONCLUSIONS

For our older patient with RA and biopsy-proven, stage IV EBER-negative HGBCL bearing MYC and BCL6 rearrangements (double hit), discontinuation of methotrexate led to a rapid and sustained marked response. Reducing immunosuppression should be considered for patients with LPDs associated with autoimmune conditions or immunosuppressive medications, regardless of additional multiagent systemic therapy administration. In older patients who are frail with aggressive B-cell lymphomas, a short trial of methotrexate withdrawal with quick interval imaging is a reasonable frontline option, regardless of EBER status.

References

1. Sesques P, Johnson NA. Approach to the diagnosis and treatment of high-grade B-cell lymphomas with MYC and BCL2 and/or BCL6 rearrangements. Blood. 2017;129(3):280-288. doi:10.1182/blood-2016-02-636316

2. Aukema SM, Siebert R, Schuuring E, et al. Double-hit B-cell lymphomas. Blood. 2011;117(8):2319-2331. doi:10.1182/blood-2010-09-297879

3. Scott DW, King RL, Staiger AM, et al. High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma morphology. Blood. 2018;131(18):2060-2064. doi:10.1182/blood-2017-12-820605

4. Dunleavy K, Fanale MA, Abramson JS, et al. Dose-adjusted EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) in untreated aggressive diffuse large B-cell lymphoma with MYC rearrangement: a prospective, multicentre, single-arm phase 2 study. Lancet Haematol. 2018;5(12):e609-e617. doi:10.1016/S2352-3026(18)30177-7

5. Younes A, Sehn LH, Johnson P, et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol. 2019;37(15):1285-1295. doi:10.1200/JCO.18.02403

6. Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood. 2021;137(5):600-609. doi:10.1182/blood.2020006578

7. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). B-cell lymphomas. Version 2.2024. January 18, 2024. Accessed January 24, 2024. https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf

8. Abbas F, Kossi ME, Shaheen IS, Sharma A, Halawa A. Post-transplantation lymphoproliferative disorders: current concepts and future therapeutic approaches. World J Transplant. 2020;10(2):29-46. doi:10.5500/wjt.v10.i2.29

9. Hoshida Y, Xu JX, Fujita S, et al. Lymphoproliferative disorders in rheumatoid arthritis: clinicopathological analysis of 76 cases in relation to methotrexate medication. J Rheumatol. 2007;34(2):322-331.

10. Salloum E, Cooper DL, Howe G, et al. Spontaneous regression of lymphoproliferative disorders in patients treated with methotrexate for rheumatoid arthritis and other rheumatic diseases. J Clin Oncol. 1996;14(6):1943-1949. doi:10.1200/JCO.1996.14.6.1943

11. Nijland ML, Kersten MJ, Pals ST, Bemelman FJ, Ten Berge IJM. Epstein-Barr virus–positive posttransplant lymphoproliferative disease after solid organ transplantation: pathogenesis, clinical manifestations, diagnosis, and management. Transplantation Direct. 2015;2(1):e48. doi:10.1097/txd.0000000000000557

12. Ekström Smedby K, Vajdic CM, Falster M, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. Blood. 2008;111(8):4029-4038. doi:10.1182/blood-2007-10-11997413. Tokuyama K, Okada F, Matsumoto S, et al. EBV-positive methotrexate-diffuse large B cell lymphoma in a rheumatoid arthritis patient. Jpn J Radiol. 2014;32(3):183-187. doi:10.1007/s11604-013-0280-y

14. Liu H, Xu-Monette ZY, Tang G, et al. EBV+ high-grade B cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements: a multi-institutional study. Histopathology. 2022;80(3):575-588. doi:10.1111/his.14585

Article PDF
Author and Disclosure Information

Nhi Nai, DOa,b; Brittany B. Coffman, MDb; Kimberly Reiter, MDb; George Atweh, MDb,c; Vishal Vashistha, MDb,c

Correspondence:  Vishal Vashistha  (vishal.vashistha@va.gov)

aUniversity of New Mexico Hospital, Department of Internal Medicine, Albuquerque

bRaymond G. Murphy New Mexico Veterans Affairs Medical Center, Albuquerque

cUniversity of New Mexico Cancer Center, Albuquerque

Author disclosures

The authors report no actual or potential conflicts of interest or outside soruces 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 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

No informed consent was obtained from the patient; patient identifiers were removed to protect the patient’s identity.

Issue
Federal Practitioner - 41(5)s
Publications
Topics
Page Number
S9
Sections
Author and Disclosure Information

Nhi Nai, DOa,b; Brittany B. Coffman, MDb; Kimberly Reiter, MDb; George Atweh, MDb,c; Vishal Vashistha, MDb,c

Correspondence:  Vishal Vashistha  (vishal.vashistha@va.gov)

aUniversity of New Mexico Hospital, Department of Internal Medicine, Albuquerque

bRaymond G. Murphy New Mexico Veterans Affairs Medical Center, Albuquerque

cUniversity of New Mexico Cancer Center, Albuquerque

Author disclosures

The authors report no actual or potential conflicts of interest or outside soruces 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 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

No informed consent was obtained from the patient; patient identifiers were removed to protect the patient’s identity.

Author and Disclosure Information

Nhi Nai, DOa,b; Brittany B. Coffman, MDb; Kimberly Reiter, MDb; George Atweh, MDb,c; Vishal Vashistha, MDb,c

Correspondence:  Vishal Vashistha  (vishal.vashistha@va.gov)

aUniversity of New Mexico Hospital, Department of Internal Medicine, Albuquerque

bRaymond G. Murphy New Mexico Veterans Affairs Medical Center, Albuquerque

cUniversity of New Mexico Cancer Center, Albuquerque

Author disclosures

The authors report no actual or potential conflicts of interest or outside soruces 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 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

No informed consent was obtained from the patient; patient identifiers were removed to protect the patient’s identity.

Article PDF
Article PDF

High-grade B-cell lymphomas (HGBCLs) are aggressive lymphoproliferative disorders (LPDs) that require fluorescence in-situ hybridization to identify gene rearrangements within MYC and BCL2 and/or BCL6 oncogenes. Traditionally referred to as double-hit or triple-hit lymphomas, HGBCL is a newer entity in the 2016 updated World Health Organization classification of lymphoid neoplasms.1 More than 90% of patients with HGBCL present with advanced clinical features, such as central nervous system involvement, leukocytosis, or lactose dehydrogenase (LDH) greater than 3 times the upper limit of normal. Treatment outcomes with aggressive multiagent chemotherapy combined with anti-CD20–targeted therapy are generally worse for patients with double-hit disease, especially among frail patients with advanced age. Patients with underlying autoimmune and rheumatologic conditions, such as rheumatoid arthritis (RA), are at higher risk for developing LPDs. These include highly aggressive subtypes of non-Hodgkin lymphoma, such as HGBCL, likely due to cascading events secondary to chronic inflammation and/or immunosuppressive medications. These immunodeficiency-associated LPDs often express positivity for Epstein-Barr virus-encoded small RNA (EBER).

We present a case of double-hit HGBCL that was EBER negative with MYC and BCL6 rearrangements in an older veteran with RA managed with methotrexate. An excellent sustained response was observed for the patient’s stage IV double-hit HGBCL disease within 4 weeks of methotrexate discontinuation. To our knowledge, this is the first reported response to methotrexate discontinuation for a patient with HGBCL.

CASE PRESENTATION

A male veteran aged 81 years presented to the Raymond G. Murphy Veterans Affairs Medical Center (RGMVAMC) in Albuquerque, New Mexico, with an unintentional 25-pound weight loss over 18 months. Pertinent history included RA managed with methotrexate 15 mg weekly for 6 years and a previous remote seizure. The patients prior prostate cancer was treated with radiation at the time of diagnosis and ongoing androgen deprivation therapy. Initial workup with chest X-ray and chest computed tomography (CT) indicated loculated left pleural fluid collection with a suspected splenic tumor.

figure_1.png

A positron-emission tomography (PET)/CT was ordered given his history of prostate cancer, which showed potential splenic and sternal metastases with corresponding fludeoxyglucose F18 uptake (Figure 1A). Biopsy was not pursued due to the potential for splenic hemorrhage. Based on the patient’s RA and methotrexate use, the collection of findings was initially thought to represent a non-Hodgkin lymphoma, with knowledge that metastatic prostate cancer refractory to androgen deprivation therapy was possible. Because he was unable to undergo a splenic biopsy, an observation strategy involving repeat PET/CT every 6 months was started.

The surveillance PET/CT 6 months later conveyed worsened disease burden with increased avidity in the manubrium (Figure 1B). The patient’s case was discussed at the RGMVAMC tumor board, and the recommendation was to continue with surveillance follow-up imaging because image-guided biopsy might not definitively yield a diagnosis. Repeat PET/CT3 months later indicated continued worsening of disease (Figure 1C) with a rapidly enlarging hypermetabolic mass in the manubrium that extended anteriorly into the subcutaneous tissues and encased the bilateral anterior jugular veins. On physical examination, this sternal mass had become painful and was clearly evident. Additionally, increased avidity in multiple upper abdominal and retroperitoneal lymph nodes was observed.

figure_2.png

Interventional radiology was consulted to assist with a percutaneous fine-needle aspiration of the manubrial mass, which revealed a dense aggregate of large, atypical lymphocytes confirmed to be of B-cell origin (CD20 and PAX5 positive) (Figure 2). The atypical B cells demonstrated co-expression of BCL6, BCL2, MUM1, and MYC but were negative for CD30 and EBER by in situ hybridization. The overall morphologic and immunophenotypic findings were consistent with a large B-cell lymphoma. Fluorescent in-situ hybridization identified the presence of MYC and BCL6 gene rearrangements, and the mass was consequently best classified as a double-hit HGBCL.

Given the patient’s history of long-term methotrexate use, we thought the HGBCL may have reflected an immunodeficiency-associated LPD, although the immunophenotype was not classic because of the CD30 and EBER negativity. With the known toxicity and poor treatment outcomes of aggressive multiagent chemotherapy for patients with double-hit HGBCL—particularly in the older adult population—methotrexate was discontinued on a trial basis.

A PET/CT was completed 4 weeks after methotrexate was discontinued due to concerns about managing an HGBCL without chemotherapy or anti-CD20–directed therapy. The updated PET/CT showed significant improvement with marked reduction in avidity of his manubrial lesion (Figure 1D). Three months after methotrexate discontinuation, the patient remained in partial remission for his double-hit HGBCL, as evidenced by no findings of sternal mass on repeat examinations with continued decrease in hypermetabolic findings on PET/CT. The patient's RA symptoms rebounded, and rheumatology colleagues prescribed sulfasalazine and periodic steroid tapers to help control his inflammatory arthritis. Fourteen months after discontinuation of methotrexate, the patient died after developing pneumonia, which led to multisystemic organ failure.

 

 

DISCUSSION

HGBCL with MYC and BCL2 and/or BCL6 rearrangements is an aggressive LPD.1 A definitive diagnosis requires collection of morphologic and immunophenotypic evaluations of suspicious tissue. Approximately 60% of patients with HGBCL have translocations in MYC and BCL2, 20% have MYC and BCL6 translocations, and the remaining 20% have MYC, BCL2 and BCL6 translocations (triple-hit disease).1

The MYC and BCL gene rearrangements are thought to synergistically drive tumorigenesis, leading to accelerated lymphoma progression and a lesser response to standard multiagent chemotherapy than seen in diffuse large B-cell lymphoma.1-3 Consequently, there have been several attempts to increase treatment efficacy with intense chemotherapy regimens, namely DA-EPOCH-R (dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab), or by adding targeted agents, such as ibrutinib and venetoclax to a standard R-CHOP (rituximab with reduced cyclophosphamide, doxorubicin, vincristine, and prednisone) backbone.4-7 Though the standard choice of therapy for fit patients harboring HGBCL remains controversial, these aggressive regimens at standard doses are typically difficult to tolerate for patients aged > 80 years.

table.png

Patients with immunosuppression are at higher risk for developing LPDs, including aggressive B-cell non-Hodgkin lymphomas such as diffuse large B-cell lymphoma. These patients are frequently classified into 2 groups: those with underlying autoimmune conditions (RA-associated LPDs), or those who have undergone solid-organ or allogeneic hematopoietic stem-cell transplants, which drives the development of posttransplant LPDs (Table).8-11 Both types of LPDs are often EBER positive, indicating some association with Epstein-Barr virus infection driven by ongoing immunosuppression, with knowledge that this finding is not absolute and is less frequent among patients with autoimmune conditions than those with posttransplant LPD.8,12

For indolent and early-stage aggressive LPDs, reduction of immunosuppression is a reasonable frontline treatment. In fact, Tokuyama and colleagues reported a previous case in which an methotrexate-associated EBER-positive early-stage diffuse large B-cell lymphoma responded well to methotrexate withdrawal.13 For advanced, aggressive LPDs associated with immunosuppression, a combination strategy of reducing immunosuppression and initiating a standard multiagent systemic therapy such as with R-CHOP is more common. Reducing immunosuppression without adding systemic anticancer therapy can certainly be considered in patients with EBER-negative LPDs; however, there is less evidence supporting this approach in the literature.

A case series of patients with EBER-positive double-hit HGBCL has been described previously, and response rates were low despite aggressive treatment.14 The current case differs from that case series in 2 ways. First, our patient did not have EBER-positive disease despite having an HGBCL associated with RA and methotrexate use. Second, our patient had a very rapid and excellent partial response simply with methotrexate discontinuation. Aggressive treatment was considered initially; however, given the patient’s age and performance status, reduction of immunosuppression alone was considered the frontline approach.

This case indicates that methotrexate withdrawal may lead to remission in patients with double-hit lymphoma, even without clear signs of Epstein-Barr virus infection being present. We are not sure why our patient with EBER-negative HGBCL responded differently to methotrexate withdrawal than the patients in the aforementioned case series with EBER-positive disease; nevertheless, a short trial of methotrexate withdrawal with repeat imaging 4 to 8 weeks after discontinuation seems reasonable for patients who are older, frail, and seemingly not fit for more aggressive treatment.

CONCLUSIONS

For our older patient with RA and biopsy-proven, stage IV EBER-negative HGBCL bearing MYC and BCL6 rearrangements (double hit), discontinuation of methotrexate led to a rapid and sustained marked response. Reducing immunosuppression should be considered for patients with LPDs associated with autoimmune conditions or immunosuppressive medications, regardless of additional multiagent systemic therapy administration. In older patients who are frail with aggressive B-cell lymphomas, a short trial of methotrexate withdrawal with quick interval imaging is a reasonable frontline option, regardless of EBER status.

High-grade B-cell lymphomas (HGBCLs) are aggressive lymphoproliferative disorders (LPDs) that require fluorescence in-situ hybridization to identify gene rearrangements within MYC and BCL2 and/or BCL6 oncogenes. Traditionally referred to as double-hit or triple-hit lymphomas, HGBCL is a newer entity in the 2016 updated World Health Organization classification of lymphoid neoplasms.1 More than 90% of patients with HGBCL present with advanced clinical features, such as central nervous system involvement, leukocytosis, or lactose dehydrogenase (LDH) greater than 3 times the upper limit of normal. Treatment outcomes with aggressive multiagent chemotherapy combined with anti-CD20–targeted therapy are generally worse for patients with double-hit disease, especially among frail patients with advanced age. Patients with underlying autoimmune and rheumatologic conditions, such as rheumatoid arthritis (RA), are at higher risk for developing LPDs. These include highly aggressive subtypes of non-Hodgkin lymphoma, such as HGBCL, likely due to cascading events secondary to chronic inflammation and/or immunosuppressive medications. These immunodeficiency-associated LPDs often express positivity for Epstein-Barr virus-encoded small RNA (EBER).

We present a case of double-hit HGBCL that was EBER negative with MYC and BCL6 rearrangements in an older veteran with RA managed with methotrexate. An excellent sustained response was observed for the patient’s stage IV double-hit HGBCL disease within 4 weeks of methotrexate discontinuation. To our knowledge, this is the first reported response to methotrexate discontinuation for a patient with HGBCL.

CASE PRESENTATION

A male veteran aged 81 years presented to the Raymond G. Murphy Veterans Affairs Medical Center (RGMVAMC) in Albuquerque, New Mexico, with an unintentional 25-pound weight loss over 18 months. Pertinent history included RA managed with methotrexate 15 mg weekly for 6 years and a previous remote seizure. The patients prior prostate cancer was treated with radiation at the time of diagnosis and ongoing androgen deprivation therapy. Initial workup with chest X-ray and chest computed tomography (CT) indicated loculated left pleural fluid collection with a suspected splenic tumor.

figure_1.png

A positron-emission tomography (PET)/CT was ordered given his history of prostate cancer, which showed potential splenic and sternal metastases with corresponding fludeoxyglucose F18 uptake (Figure 1A). Biopsy was not pursued due to the potential for splenic hemorrhage. Based on the patient’s RA and methotrexate use, the collection of findings was initially thought to represent a non-Hodgkin lymphoma, with knowledge that metastatic prostate cancer refractory to androgen deprivation therapy was possible. Because he was unable to undergo a splenic biopsy, an observation strategy involving repeat PET/CT every 6 months was started.

The surveillance PET/CT 6 months later conveyed worsened disease burden with increased avidity in the manubrium (Figure 1B). The patient’s case was discussed at the RGMVAMC tumor board, and the recommendation was to continue with surveillance follow-up imaging because image-guided biopsy might not definitively yield a diagnosis. Repeat PET/CT3 months later indicated continued worsening of disease (Figure 1C) with a rapidly enlarging hypermetabolic mass in the manubrium that extended anteriorly into the subcutaneous tissues and encased the bilateral anterior jugular veins. On physical examination, this sternal mass had become painful and was clearly evident. Additionally, increased avidity in multiple upper abdominal and retroperitoneal lymph nodes was observed.

figure_2.png

Interventional radiology was consulted to assist with a percutaneous fine-needle aspiration of the manubrial mass, which revealed a dense aggregate of large, atypical lymphocytes confirmed to be of B-cell origin (CD20 and PAX5 positive) (Figure 2). The atypical B cells demonstrated co-expression of BCL6, BCL2, MUM1, and MYC but were negative for CD30 and EBER by in situ hybridization. The overall morphologic and immunophenotypic findings were consistent with a large B-cell lymphoma. Fluorescent in-situ hybridization identified the presence of MYC and BCL6 gene rearrangements, and the mass was consequently best classified as a double-hit HGBCL.

Given the patient’s history of long-term methotrexate use, we thought the HGBCL may have reflected an immunodeficiency-associated LPD, although the immunophenotype was not classic because of the CD30 and EBER negativity. With the known toxicity and poor treatment outcomes of aggressive multiagent chemotherapy for patients with double-hit HGBCL—particularly in the older adult population—methotrexate was discontinued on a trial basis.

A PET/CT was completed 4 weeks after methotrexate was discontinued due to concerns about managing an HGBCL without chemotherapy or anti-CD20–directed therapy. The updated PET/CT showed significant improvement with marked reduction in avidity of his manubrial lesion (Figure 1D). Three months after methotrexate discontinuation, the patient remained in partial remission for his double-hit HGBCL, as evidenced by no findings of sternal mass on repeat examinations with continued decrease in hypermetabolic findings on PET/CT. The patient's RA symptoms rebounded, and rheumatology colleagues prescribed sulfasalazine and periodic steroid tapers to help control his inflammatory arthritis. Fourteen months after discontinuation of methotrexate, the patient died after developing pneumonia, which led to multisystemic organ failure.

 

 

DISCUSSION

HGBCL with MYC and BCL2 and/or BCL6 rearrangements is an aggressive LPD.1 A definitive diagnosis requires collection of morphologic and immunophenotypic evaluations of suspicious tissue. Approximately 60% of patients with HGBCL have translocations in MYC and BCL2, 20% have MYC and BCL6 translocations, and the remaining 20% have MYC, BCL2 and BCL6 translocations (triple-hit disease).1

The MYC and BCL gene rearrangements are thought to synergistically drive tumorigenesis, leading to accelerated lymphoma progression and a lesser response to standard multiagent chemotherapy than seen in diffuse large B-cell lymphoma.1-3 Consequently, there have been several attempts to increase treatment efficacy with intense chemotherapy regimens, namely DA-EPOCH-R (dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab), or by adding targeted agents, such as ibrutinib and venetoclax to a standard R-CHOP (rituximab with reduced cyclophosphamide, doxorubicin, vincristine, and prednisone) backbone.4-7 Though the standard choice of therapy for fit patients harboring HGBCL remains controversial, these aggressive regimens at standard doses are typically difficult to tolerate for patients aged > 80 years.

table.png

Patients with immunosuppression are at higher risk for developing LPDs, including aggressive B-cell non-Hodgkin lymphomas such as diffuse large B-cell lymphoma. These patients are frequently classified into 2 groups: those with underlying autoimmune conditions (RA-associated LPDs), or those who have undergone solid-organ or allogeneic hematopoietic stem-cell transplants, which drives the development of posttransplant LPDs (Table).8-11 Both types of LPDs are often EBER positive, indicating some association with Epstein-Barr virus infection driven by ongoing immunosuppression, with knowledge that this finding is not absolute and is less frequent among patients with autoimmune conditions than those with posttransplant LPD.8,12

For indolent and early-stage aggressive LPDs, reduction of immunosuppression is a reasonable frontline treatment. In fact, Tokuyama and colleagues reported a previous case in which an methotrexate-associated EBER-positive early-stage diffuse large B-cell lymphoma responded well to methotrexate withdrawal.13 For advanced, aggressive LPDs associated with immunosuppression, a combination strategy of reducing immunosuppression and initiating a standard multiagent systemic therapy such as with R-CHOP is more common. Reducing immunosuppression without adding systemic anticancer therapy can certainly be considered in patients with EBER-negative LPDs; however, there is less evidence supporting this approach in the literature.

A case series of patients with EBER-positive double-hit HGBCL has been described previously, and response rates were low despite aggressive treatment.14 The current case differs from that case series in 2 ways. First, our patient did not have EBER-positive disease despite having an HGBCL associated with RA and methotrexate use. Second, our patient had a very rapid and excellent partial response simply with methotrexate discontinuation. Aggressive treatment was considered initially; however, given the patient’s age and performance status, reduction of immunosuppression alone was considered the frontline approach.

This case indicates that methotrexate withdrawal may lead to remission in patients with double-hit lymphoma, even without clear signs of Epstein-Barr virus infection being present. We are not sure why our patient with EBER-negative HGBCL responded differently to methotrexate withdrawal than the patients in the aforementioned case series with EBER-positive disease; nevertheless, a short trial of methotrexate withdrawal with repeat imaging 4 to 8 weeks after discontinuation seems reasonable for patients who are older, frail, and seemingly not fit for more aggressive treatment.

CONCLUSIONS

For our older patient with RA and biopsy-proven, stage IV EBER-negative HGBCL bearing MYC and BCL6 rearrangements (double hit), discontinuation of methotrexate led to a rapid and sustained marked response. Reducing immunosuppression should be considered for patients with LPDs associated with autoimmune conditions or immunosuppressive medications, regardless of additional multiagent systemic therapy administration. In older patients who are frail with aggressive B-cell lymphomas, a short trial of methotrexate withdrawal with quick interval imaging is a reasonable frontline option, regardless of EBER status.

References

1. Sesques P, Johnson NA. Approach to the diagnosis and treatment of high-grade B-cell lymphomas with MYC and BCL2 and/or BCL6 rearrangements. Blood. 2017;129(3):280-288. doi:10.1182/blood-2016-02-636316

2. Aukema SM, Siebert R, Schuuring E, et al. Double-hit B-cell lymphomas. Blood. 2011;117(8):2319-2331. doi:10.1182/blood-2010-09-297879

3. Scott DW, King RL, Staiger AM, et al. High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma morphology. Blood. 2018;131(18):2060-2064. doi:10.1182/blood-2017-12-820605

4. Dunleavy K, Fanale MA, Abramson JS, et al. Dose-adjusted EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) in untreated aggressive diffuse large B-cell lymphoma with MYC rearrangement: a prospective, multicentre, single-arm phase 2 study. Lancet Haematol. 2018;5(12):e609-e617. doi:10.1016/S2352-3026(18)30177-7

5. Younes A, Sehn LH, Johnson P, et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol. 2019;37(15):1285-1295. doi:10.1200/JCO.18.02403

6. Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood. 2021;137(5):600-609. doi:10.1182/blood.2020006578

7. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). B-cell lymphomas. Version 2.2024. January 18, 2024. Accessed January 24, 2024. https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf

8. Abbas F, Kossi ME, Shaheen IS, Sharma A, Halawa A. Post-transplantation lymphoproliferative disorders: current concepts and future therapeutic approaches. World J Transplant. 2020;10(2):29-46. doi:10.5500/wjt.v10.i2.29

9. Hoshida Y, Xu JX, Fujita S, et al. Lymphoproliferative disorders in rheumatoid arthritis: clinicopathological analysis of 76 cases in relation to methotrexate medication. J Rheumatol. 2007;34(2):322-331.

10. Salloum E, Cooper DL, Howe G, et al. Spontaneous regression of lymphoproliferative disorders in patients treated with methotrexate for rheumatoid arthritis and other rheumatic diseases. J Clin Oncol. 1996;14(6):1943-1949. doi:10.1200/JCO.1996.14.6.1943

11. Nijland ML, Kersten MJ, Pals ST, Bemelman FJ, Ten Berge IJM. Epstein-Barr virus–positive posttransplant lymphoproliferative disease after solid organ transplantation: pathogenesis, clinical manifestations, diagnosis, and management. Transplantation Direct. 2015;2(1):e48. doi:10.1097/txd.0000000000000557

12. Ekström Smedby K, Vajdic CM, Falster M, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. Blood. 2008;111(8):4029-4038. doi:10.1182/blood-2007-10-11997413. Tokuyama K, Okada F, Matsumoto S, et al. EBV-positive methotrexate-diffuse large B cell lymphoma in a rheumatoid arthritis patient. Jpn J Radiol. 2014;32(3):183-187. doi:10.1007/s11604-013-0280-y

14. Liu H, Xu-Monette ZY, Tang G, et al. EBV+ high-grade B cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements: a multi-institutional study. Histopathology. 2022;80(3):575-588. doi:10.1111/his.14585

References

1. Sesques P, Johnson NA. Approach to the diagnosis and treatment of high-grade B-cell lymphomas with MYC and BCL2 and/or BCL6 rearrangements. Blood. 2017;129(3):280-288. doi:10.1182/blood-2016-02-636316

2. Aukema SM, Siebert R, Schuuring E, et al. Double-hit B-cell lymphomas. Blood. 2011;117(8):2319-2331. doi:10.1182/blood-2010-09-297879

3. Scott DW, King RL, Staiger AM, et al. High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements with diffuse large B-cell lymphoma morphology. Blood. 2018;131(18):2060-2064. doi:10.1182/blood-2017-12-820605

4. Dunleavy K, Fanale MA, Abramson JS, et al. Dose-adjusted EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) in untreated aggressive diffuse large B-cell lymphoma with MYC rearrangement: a prospective, multicentre, single-arm phase 2 study. Lancet Haematol. 2018;5(12):e609-e617. doi:10.1016/S2352-3026(18)30177-7

5. Younes A, Sehn LH, Johnson P, et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol. 2019;37(15):1285-1295. doi:10.1200/JCO.18.02403

6. Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. Blood. 2021;137(5):600-609. doi:10.1182/blood.2020006578

7. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). B-cell lymphomas. Version 2.2024. January 18, 2024. Accessed January 24, 2024. https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf

8. Abbas F, Kossi ME, Shaheen IS, Sharma A, Halawa A. Post-transplantation lymphoproliferative disorders: current concepts and future therapeutic approaches. World J Transplant. 2020;10(2):29-46. doi:10.5500/wjt.v10.i2.29

9. Hoshida Y, Xu JX, Fujita S, et al. Lymphoproliferative disorders in rheumatoid arthritis: clinicopathological analysis of 76 cases in relation to methotrexate medication. J Rheumatol. 2007;34(2):322-331.

10. Salloum E, Cooper DL, Howe G, et al. Spontaneous regression of lymphoproliferative disorders in patients treated with methotrexate for rheumatoid arthritis and other rheumatic diseases. J Clin Oncol. 1996;14(6):1943-1949. doi:10.1200/JCO.1996.14.6.1943

11. Nijland ML, Kersten MJ, Pals ST, Bemelman FJ, Ten Berge IJM. Epstein-Barr virus–positive posttransplant lymphoproliferative disease after solid organ transplantation: pathogenesis, clinical manifestations, diagnosis, and management. Transplantation Direct. 2015;2(1):e48. doi:10.1097/txd.0000000000000557

12. Ekström Smedby K, Vajdic CM, Falster M, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. Blood. 2008;111(8):4029-4038. doi:10.1182/blood-2007-10-11997413. Tokuyama K, Okada F, Matsumoto S, et al. EBV-positive methotrexate-diffuse large B cell lymphoma in a rheumatoid arthritis patient. Jpn J Radiol. 2014;32(3):183-187. doi:10.1007/s11604-013-0280-y

14. Liu H, Xu-Monette ZY, Tang G, et al. EBV+ high-grade B cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements: a multi-institutional study. Histopathology. 2022;80(3):575-588. doi:10.1111/his.14585

Issue
Federal Practitioner - 41(5)s
Issue
Federal Practitioner - 41(5)s
Page Number
S9
Page Number
S9
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>0524 epub AVAHO Methotrexate</fileName> <TBEID>0C02F2D9.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F2D9</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240408T160515</firstPublished> <LastPublished>20240428T215720</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240408T160514</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Nhi Nai, DOa,b; Brittany B. Coffman, MDb; Kimberly Reiter, MDb; George Atweh, MDb,c; Vishal Vashistha, MDb,c</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>H igh-grade B-cell lymphomas (HGBCLs) are aggressive lymphoproliferative disorders (LPDs) that require fluorescence in-situ hybridization to identify gene rearr</metaDescription> <articlePDF/> <teaserImage/> <title>EBER-Negative, Double-Hit High-Grade B-Cell Lymphoma Responding to Methotrexate Discontinuation</title> <deck/> <eyebrow>Case in Point</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>2</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2953</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case in Point | 2953<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>112</term> <term canonical="true">45</term> </sections> <topics> <term canonical="true">28798</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>EBER-Negative, Double-Hit High-Grade B-Cell Lymphoma Responding to Methotrexate Discontinuation</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> First classified in 2016, high-grade B-cell lymphoma (HGBCL) is a lymphoid neoplasm that is typically seen as an aggressive lymphoproliferative disorder (LPD). In most patients with HGBCL, various oncogene rearrangements present with advanced clinical features, such as central nervous system involvement. Patients with underlying autoimmune and rheumatologic conditions, such as rheumatoid arthritis, are at higher risk for developing LPDs, including highly aggressive subtypes of non-Hodgkin lymphomas such as HGBCL.<br/><br/><b>Case Presentation: </b>We present a case of stage IV double-hit HGBCL with the presence of <i>MYC</i> and <i>BCL6</i> gene rearrangements in an older veteran with rheumatoid arthritis treated with methotrexate. An excellent sustained response was observed for the patient’s disease within 4 weeks of methotrexate discontinuation. To our knowledge, this is the first reported response to methotrexate discontinuation for a patient with HGBCL.<br/><br/><b>Conclusions:</b> Reducing immunosuppression should be considered in all patients with LPDs associated with autoimmune conditions or immunosuppressive medications, regardless of additional multiagent systemic therapy administration. </p> <p>H igh-grade B-cell lymphomas (HGBCLs) are aggressive lymphoproliferative disorders (LPDs) that require fluorescence in-situ hybridization to identify gene rearrangements within <i>MYC</i> and <i>BCL2</i> and/or <i>BCL6</i> oncogenes. Traditionally referred to as double-hit or triple-hit lymphomas, HGBCL is a newer entity in the 2016 updated World Health Organization classification of lymphoid neoplasms.<sup>1</sup> More than 90% of patients with HGBCL present with advanced clinical features, such as central nervous system involvement, leukocytosis, or lactose dehydrogenase (LDH) greater than 3 times the upper limit of normal. Treatment outcomes with aggressive multiagent chemotherapy combined with anti-CD20–targeted therapy are generally worse for patients with double-hit disease, especially among frail patients with advanced age. Patients with underlying autoimmune and rheumatologic conditions, such as rheumatoid arthritis (RA), are at higher risk for developing LPDs. These include highly aggressive subtypes of non-Hodgkin lymphoma, such as HGBCL, likely due to cascading events secondary to chronic inflammation and/or immunosuppressive medications. These immunodeficiency-associated LPDs often express positivity for Epstein-Barr virus-encoded small RNA (EBER).</p> <p>We present a case of double-hit HGBCL that was EBER negative with <i>MYC</i> and <i>BCL6</i> rearrangements in an older veteran with RA managed with methotrexate. An excellent sustained response was observed for the patient’s stage IV double-hit HGBCL disease within 4 weeks of methotrexate discontinuation. To our knowledge, this is the first reported response to methotrexate discontinuation for a patient with HGBCL. </p> <h2>CASE PRESENTATION</h2> <p>A male veteran aged 81 years presented to the Raymond G. Murphy Veterans Affairs Medical Center (RGMVAMC) in Albuquerque, New Mexico, with an unintentional 25-pound weight loss over 18 months. Pertinent history included RA managed with methotrexate 15 mg weekly for 6 years and a previous remote seizure. The patients prior prostate cancer was treated with radiation at the time of diagnosis and ongoing androgen deprivation therapy. Initial workup with chest X-ray and chest computed tomography (CT) indicated loculated left pleural fluid collection with a suspected splenic tumor. </p> <p>A positron-emission tomography (PET)/CT was ordered given his history of prostate cancer, which showed potential splenic and sternal metastases with corresponding fludeoxyglucose F18 uptake (Figure 1A). Biopsy was not pursued due to the potential for splenic hemorrhage. Based on the patient’s RA and methotrexate use, the collection of findings was initially thought to represent a non-Hodgkin lymphoma, with knowledge that metastatic prostate cancer refractory to androgen deprivation therapy was possible. Because he was unable to undergo a splenic biopsy, an observation strategy involving repeat PET/CT every 6 months was started.<br/><br/>The surveillance PET/CT 6 months later conveyed worsened disease burden with increased avidity in the manubrium (Figure 1B). The patient’s case was discussed at the RGMVAMC tumor board, and the recommendation was to continue with surveillance follow-up imaging because image-guided biopsy might not definitively yield a diagnosis. Repeat PET/CT3 months later indicated continued worsening of disease (Figure 1C) with a rapidly enlarging hypermetabolic mass in the manubrium that extended anteriorly into the subcutaneous tissues and encased the bilateral anterior jugular veins. On physical examination, this sternal mass had become painful and was clearly evident. Additionally, increased avidity in multiple upper abdominal and retroperitoneal lymph nodes was observed.Interventional radiology was consulted to assist with a percutaneous fine-needle aspiration of the manubrial mass, which revealed a dense aggregate of large, atypical lymphocytes confirmed to be of B-cell origin (CD20 and PAX5 positive) (Figure 2). The atypical B cells demonstrated co-expression of <i>BCL6</i>, <i>BCL2</i>, <i>MUM1</i>, and <i>MYC</i> but were negative for CD30 and EBER by in situ hybridization. The overall morphologic and immunophenotypic findings were consistent with a large B-cell lymphoma. Fluorescent in-situ hybridization identified the presence of <i>MYC</i> and <i>BCL6</i> gene rearrangements, and the mass was consequently best classified as a double-hit HGBCL. <br/><br/>Given the patient’s history of long-term methotrexate use, we thought the HGBCL may have reflected an immunodeficiency-associated LPD, although the immunophenotype was not classic because of the CD30 and EBER negativity. With the known toxicity and poor treatment outcomes of aggressive multiagent chemotherapy for patients with double-hit HGBCL—particularly in the older adult population—methotrexate was discontinued on a trial basis. <br/><br/>A PET/CT was completed 4 weeks after methotrexate was discontinued due to concerns about managing an HGBCL without chemotherapy or anti-CD20–directed therapy. The updated PET/CT showed significant improvement with marked reduction in avidity of his manubrial lesion (Figure 1D). Three months after methotrexate discontinuation, the patient remained in partial remission for his double-hit HGBCL, as evidenced by no findings of sternal mass on repeat examinations with continued decrease in hypermetabolic findings on PET/CT. The patient's RA symptoms rebounded, and rheumatology colleagues prescribed sulfasalazine and periodic steroid tapers to help control his inflammatory arthritis. Fourteen months after discontinuation of methotrexate, the patient died after developing pneumonia, which led to multisystemic organ failure.</p> <h2>DISCUSSION</h2> <p>HGBCL with <i>MYC </i>and <i>BCL2 </i>and/or <i>BCL6</i> rearrangements is an aggressive LPD.<sup>1</sup> A definitive diagnosis requires collection of morphologic and immunophenotypic evaluations of suspicious tissue. Approximately 60% of patients with HGBCL have translocations in <i>MYC</i> and <i>BCL2</i>, 20% have <i>MYC </i>and <i>BCL6</i> translocations, and the remaining 20% have <i>MYC</i>, <i>BCL2 </i>and <i>BCL6</i> translocations (triple-hit disease).<sup>1</sup> </p> <p>The <i>MYC and BCL</i> gene rearrangements are thought to synergistically drive tumorigenesis, leading to accelerated lymphoma progression and a lesser response to standard multiagent chemotherapy than seen in diffuse large B-cell lymphoma.<sup>1-3</sup> Consequently, there have been several attempts to increase treatment efficacy with intense chemotherapy regimens, namely DA-EPOCH-R (dose-adjusted etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab), or by adding targeted agents, such as ibrutinib and venetoclax to a standard R-CHOP (rituximab with reduced cyclophosphamide, doxorubicin, vincristine, and prednisone) backbone.<sup>4-7</sup> Though the standard choice of therapy for fit patients harboring HGBCL remains controversial, these aggressive regimens at standard doses are typically difficult to tolerate for patients aged &gt; 80 years.<br/><br/>Patients with immunosuppression are at higher risk for developing LPDs, including aggressive B-cell non-Hodgkin lymphomas such as diffuse large B-cell lymphoma. These patients are frequently classified into 2 groups: those with underlying autoimmune conditions (RA-associated LPDs), or those who have undergone solid-organ or allogeneic hematopoietic stem-cell transplants, which drives the development of posttransplant LPDs (Table).<sup>8-11</sup> Both types of LPDs are often EBER positive, indicating some association with Epstein-Barr virus infection driven by ongoing immunosuppression, with knowledge that this finding is not absolute and is less frequent among patients with autoimmune conditions than those with posttransplant LPD.<sup>8,12</sup> <br/><br/>For indolent and early-stage aggressive LPDs, reduction of immunosuppression is a reasonable frontline treatment. In fact, Tokuyama and colleagues reported a previous case in which an methotrexate-associated EBER-positive early-stage diffuse large B-cell lymphoma responded well to methotrexate withdrawal.<sup>13</sup> For advanced, aggressive LPDs associated with immunosuppression, a combination strategy of reducing immunosuppression and initiating a standard multiagent systemic therapy such as with R-CHOP is more common. Reducing immunosuppression without adding systemic anticancer therapy can certainly be considered in patients with EBER-negative LPDs; however, there is less evidence supporting this approach in the literature. <br/><br/>A case series of patients with EBER-positive double-hit HGBCL has been described previously, and response rates were low despite aggressive treatment.<sup>14</sup> The current case differs from that case series in 2 ways. First, our patient did not have EBER-positive disease despite having an HGBCL associated with RA and methotrexate use. Second, our patient had a very rapid and excellent partial response simply with methotrexate discontinuation. Aggressive treatment was considered initially; however, given the patient’s age and performance status, reduction of immunosuppression alone was considered the frontline approach. <br/><br/>This case indicates that methotrexate withdrawal may lead to remission in patients with double-hit lymphoma, even without clear signs of Epstein-Barr virus infection being present. We are not sure why our patient with EBER-negative HGBCL responded differently to methotrexate withdrawal than the patients in the aforementioned case series with EBER-positive disease; nevertheless, a short trial of methotrexate withdrawal with repeat imaging 4 to 8 weeks after discontinuation seems reasonable for patients who are older, frail, and seemingly not fit for more aggressive treatment. </p> <h2>CONCLUSIONS</h2> <p>For our older patient with RA and biopsy-proven, stage IV EBER-negative HGBCL bearing <i>MYC </i>and <i>BCL6 </i>rearrangements (double hit), discontinuation of methotrexate led to a rapid and sustained marked response. Reducing immunosuppression should be considered for patients with LPDs associated with autoimmune conditions or immunosuppressive medications, regardless of additional multiagent systemic therapy administration. In older patients who are frail with aggressive B-cell lymphomas, a short trial of methotrexate withdrawal with quick interval imaging is a reasonable frontline option, regardless of EBER status. </p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>University of New Mexico Hospital, Department of Internal Medicine, Albuquerque<br/><br/><sup>b</sup>Raymond G. Murphy New Mexico Veterans Affairs Medical Center, Albuquerque <br/><br/><sup>c</sup>University of New Mexico Cancer Center, Albuquerque</em> </p> <p class="isub">Author disclosures</p> <p> <em>The authors report no actual or potential conflicts of interest or outside soruces of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <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> <p class="isub">Ethics and consent</p> <p> <em>No informed consent was obtained from the patient; patient identifiers were removed to protect the patient’s identity.</em> </p> <h2>References</h2> <p class="reference"> 1. Sesques P, Johnson NA. Approach to the diagnosis and treatment of high-grade B-cell lymphomas with <i>MYC</i> and <i>BCL2</i> and/or <i>BCL6</i> rearrangements. <i>Blood</i>. 2017;129(3):280-288. doi:10.1182/blood-2016-02-636316 <br/><br/> 2. Aukema SM, Siebert R, Schuuring E, et al. Double-hit B-cell lymphomas. <i>Blood</i>. 2011;117(8):2319-2331. doi:10.1182/blood-2010-09-297879 <br/><br/> 3. Scott DW, King RL, Staiger AM, et al. High-grade B-cell lymphoma with <i>MYC</i> and <i>BCL2</i> and/or <i>BCL6</i> rearrangements with diffuse large B-cell lymphoma morphology. <i>Blood</i>. 2018;131(18):2060-2064. doi:10.1182/blood-2017-12-820605 <br/><br/> 4. Dunleavy K, Fanale MA, Abramson JS, et al. Dose-adjusted EPOCH-R (etoposide, prednisone, vincristine, cyclophosphamide, doxorubicin, and rituximab) in untreated aggressive diffuse large B-cell lymphoma with <i>MYC</i> rearrangement: a prospective, multicentre, single-arm phase 2 study. <i>Lancet Haematol</i>. 2018;5(12):e609-e617. doi:10.1016/S2352-3026(18)30177-7<br/><br/> 5. Younes A, Sehn LH, Johnson P, et al. Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. <i>J Clin Oncol</i>. 2019;37(15):1285-1295. doi:10.1200/JCO.18.02403<br/><br/> 6. Morschhauser F, Feugier P, Flinn IW, et al. A phase 2 study of venetoclax plus R-CHOP as first-line treatment for patients with diffuse large B-cell lymphoma. <i>Blood</i>. 2021;137(5):600-609. doi:10.1182/blood.2020006578<br/><br/> 7. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines<sup>®</sup>). B-cell lymphomas. Version 2.2024. January 18, 2024. Accessed January 24, 2024. https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf <br/><br/> 8. Abbas F, Kossi ME, Shaheen IS, Sharma A, Halawa A. Post-transplantation lymphoproliferative disorders: current concepts and future therapeutic approaches. <i>World J Transplant</i>. 2020;10(2):29-46. doi:10.5500/wjt.v10.i2.29 <br/><br/> 9. Hoshida Y, Xu JX, Fujita S, et al. Lymphoproliferative disorders in rheumatoid arthritis: clinicopathological analysis of 76 cases in relation to methotrexate medication. <i>J Rheumatol</i>. 2007;34(2):322-331. <br/><br/>10. Salloum E, Cooper DL, Howe G, et al. Spontaneous regression of lymphoproliferative disorders in patients treated with methotrexate for rheumatoid arthritis and other rheumatic diseases. <i>J Clin Oncol</i>. 1996;14(6):1943-1949. doi:10.1200/JCO.1996.14.6.1943 <br/><br/>11. Nijland ML, Kersten MJ, Pals ST, Bemelman FJ, Ten Berge IJM. Epstein-Barr virus–positive posttransplant lymphoproliferative disease after solid organ transplantation: pathogenesis, clinical manifestations, diagnosis, and management. <i>Transplantation Direct</i>. 2015;2(1):e48. doi:10.1097/txd.0000000000000557 <br/><br/>12. Ekström Smedby K, Vajdic CM, Falster M, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. <i>Blood</i>. 2008;111(8):4029-4038. doi:10.1182/blood-2007-10-11997413. Tokuyama K, Okada F, Matsumoto S, et al. EBV-positive methotrexate-diffuse large B cell lymphoma in a rheumatoid arthritis patient. <i>Jpn J Radiol</i>. 2014;32(3):183-187. doi:10.1007/s11604-013-0280-y<br/><br/>14. Liu H, Xu-Monette ZY, Tang G, et al. EBV<sup>+</sup> high-grade B cell lymphoma with <i>MYC</i> and <i>BCL2</i> and/or <i>BCL6</i> rearrangements: a multi-institutional study. <i>Histopathology</i>. 2022;80(3):575-588. doi:10.1111/his.14585 </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
Image
Disable zoom
Off

A Case of Metastatic Chromophobe Renal Cell Carcinoma Masked as Suspected Hepatic Abscesses

Article Type
Changed
Mon, 04/29/2024 - 19:50

Finding new liver lesions on imaging during a febrile illness may indicate hepatic abscesses or malignancy. These can be difficult to diagnose with imaging alone. Differentiating between infectious and neoplastic etiologies may require additional images and/or tissue samples.

Hepatic abscesses are more commonly seen with other abdominal or biliary infections while metastatic disease usually presents in patients with active cancer or on surveillance imaging. While renal cell carcinoma (RCC) is the most common malignant neoplasm of the kidney, chromophobe renal cell carcinoma (chRCC) is a rare subtype that comprises only 5% of RCC cases.1 We present a case of a patient with numerous new liver lesions and fever, initially thought to be hepatic abscesses, who was found to have metastatic chRCC.

 

CASE PRESENTATION

A 53 year-old male with a history of stage 2 chRCC and right radical nephrectomy 2 years prior presented to the emergency department following 1 week of drenching night sweats, fatigue, and subjective fevers. In addition, the patient reported gradually progressive, dull, right upper-quadrant abdominal pain. He noted no other acute medical complaints at the time of presentation. His history was notable for hyperlipidemia. His only surgery was the nephrectomy 2 years earlier. The patient reported no alcohol, tobacco, or drug use, any recent travel, or pet or animal exposure. On admission, he was afebrile with normal heart rate and was normotensive. His physical examination was remarkable for hepatomegaly with right upper-quadrant abdominal tenderness to palpation with a negative Murphy sign. There were otherwise no abnormal cardiovascular, respiratory, or skin findings.

Laboratory tests during initial evaluation were notable for hemoglobin of 10.0 g/dL, white blood cell count of 16.7×103 μL, alkaline phosphatase of 213 U/L, aspartate aminotransferase of 185 U/L, and alanine aminotransferase of 36 U/L. Screening tests for viral hepatitis A, B, and C were negative. Additional tests for HIV, rapid plasma reagin, Epstein-Barr virus, cytomegalovirus, and toxoplasma were negative. Tests for antimitochondrial, antismooth muscle, and serum antinuclear antibodies were negative.

figure_1.png

Chest X-ray did not reveal any acute cardiopulmonary process. Computed tomography with contrast of the abdomen and pelvis demonstrated numerous hypodensities within the right hepatic lobe. Right upper-quadrant ultrasound demonstrated multiple hyperechoic foci throughout the liver. confluent decreased T1 signal lesions with peripheral gadolinium hyperenhancement were evident on Gadolinium-enhanced T1-weighted magnetic resonance imaging (MRI) with fat saturation demonstrated numerous (Figure 1).

Liver biopsy and tissue cultures demonstrated normal hepatic tissue and no organism growth. Blood cultures demonstrated no growth. The patient was empirically treated with IV ceftriaxone 1 g daily and metronidazole 500 mg every 8 hours for suspected hepatic abscesses after he was admitted to the hospital.

figure_2.png

The patient’s symptoms initially improved following antibiotic treatment; however, he reported recurrence of the initial symptoms2 months later at a follow-up appointment with gastroenterology. Liver-associated enzymes also remained elevated despite 4 weeks of antibiotic treatment. Repeat gadolinium-enhanced T1 fat-saturated MRI demonstrated an interval increase in size and number of confluent hepatic lesions throughout the liver (Figure 2).

figure_4.png

figure_3.png

A repeat liver biopsy revealed metastatic chRCC (Figures 3 and 4) that was both morphologically and immunohistochemically similar to the first pathologic diagnosis made following nephrectomy. The first liver biopsy likely did not sample the metastatic lesions that were present but instead sampled the surrounding normal liver. The patient was initiated on lenvatinib and everolimus therapy with oncology, a recommended regimen per the National Comprehensive Cancer Network for patients with nonclear cell RCC.1

 

 

DISCUSSION

Chromophobe RCC is a rare form of RCC that has a recurrence-free survival of > 80% when treated in early stages.2 These neoplasms represent only 3000 to 6000 new cases of RCC annually, with an even lower incidence (6% to 7%) resulting in metastatic disease. The liver is the most common site of metastases (39%).2 Previously reported metastatic chRCC hepatic lesions have been incidentally noted on imaging with asymptomatic clinical presentations. In contrast to our patient, most documented cases report metastatic chRCC as a solitary hepatic lesion.3-7

A noteworthy genetic association with ChRCC is the Birt-Hogg-Dubé syndrome, which is an autosomal-dominant genetic disorder that results from germline mutations in the tumor suppressor folliculin gene located on chromosome 17.8 This syndrome is characterized by the development of various benign and malignant tumors, particularly chRCC. Our patient appeared to have a sporadic chRCC with the absence of other tumors and negative family history for malignancies. On his initial staging imaging, in accordance with National Comprehensive Cancer Network guidelines, our patient was identified only as having a 10-cm right renal mass and 1 benign regional lymph node with an otherwise unremarkable computed tomography of the chest, abdomen and pelvis, corresponding to stage 2 cancer.

Common causes of hepatic abscesses, the other potential diagnosis of concern for the patient, were biliary infections, portal vein ascension from abdominal sources, arterial translocation due to bacteremia, and local invasion due to suppuration of adjacent tissue.9 Incidence for hepatic abscesses increases with comorbidities such as diabetes, cirrhosis, malignancy, immunosuppression, and malnutrition.10Candida is also a common culprit when there are multiple, small abscesses, often in immunocompromised patients.11 Given the high mortality rates associated with hepatic abscesses, prompt treatment is imperative.10,12 Since the clinical signs and symptoms for hepatic abscesses are nonspecific (abdominal pain, fever, and malaise) and liver function tests can vary, the diagnosis primarily relies on imaging or tissue sampling.9

It can be difficult to distinguish abscesses from metastatic lesions based on imaging alone without microbiologic and pathologic confirmation.11,13,14 There are certain radiologic characteristics that have been found to favor abscesses over metastasis, including parenchymal enhancement, arterial rim enhancement, and perilesional hyperemia.15 However, previously described hallmark characteristics of hepatic abscesses, such as the “cluster sign” demonstrating early stages of abscess coalescence, have also been seen in some hepatic metastases.16

CONCLUSIONS

This patient highlights the presentation of a rare case of metastatic chRCC with multiple hepatic lesions. While often differentiated clinically, radiographically, or histologically, malignancy and abscess can be difficult to differentiate in a patient with fevers and leukocytosis with hepatic lesions.17 Early diagnosis of hepatic abscesses and initiation of antibiotic therapy are essential. In cases when it is challenging to characterize the hepatic lesions, repeated tissue sampling and imaging can help direct therapy. Attention should be paid to a previous history of malignancy and should raise suspicion for metastatic disease, particularly with misleading imaging studies and tissue samples.

Acknowledgments

This case was presented as a poster presentation at the Tri-Service American College of Physicians Meeting, September 7-10, 2022, San Antonio, Texas.

References

1. National Comprehensive Cancer Network. Kidney cancer (version 2.2024). Accessed February 5, 2024. https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf

2. Vera-Badillo FE, Conde E, Duran I. Chromophobe renal cell carcinoma: a review of an uncommon entity. Int J Urol. 2012;19(10):894-900.doi:10.1111/j.1442-2042.2012.03079.x

3. Lordan JT, Fawcett WJ, Karanjia ND. Solitary liver metastasis of chromophobe renal cell carcinoma 20 years after nephrectomy treated by hepatic resection. Urology. 2008;72(1):230.e5-6. doi:10.1016/j.urology.2007.11.134

4. Talarico F, Buli P, Iusco D, Sangiorgi A, Jovine E. Synchronous nephrectomy and right hepatectomy for metastatic chromophobe renal cell carcinoma: report of a case and review of the literature. Chir Ital. 2007;59(2):257-261.

5. Aslam MI, Spencer L, Garcea G, et al. A case of liver metastasis from an oncocytoma with a focal area of chromophobe renal cell carcinoma: a wolf in sheep’s clothing. Int J Surg Pathol. 2008;17(2):158-162. doi:10.1177/1066896908318741

6. Kyoda Y, Kobayashi K, Takahashi A, et al. Liver metastasis with portal vein tumor thrombosis as a late recurrence of chromophobe renal cell carcinoma. Article in Japanese. Hinyokika Kiyo. 2009;55(1):23-25.

7. Talarico F, Capizzi D, Iusco DR. Solitary liver metastasis of chromophobe renal cell carcinoma 17 years after nephrectomy. a case report and review of the literature. Ann Ital Chir. 2013;84(ePub):S2239253X13021816.

8. Garje R, Elhag D, Yasin HA, Acharya L, Vaena D, Dahmoush L. Comprehensive review of chromophobe renal cell carcinoma. Crit Rev Oncol Hematol. 2021;160:103287. doi:10.1016/j.critrevonc.2021.103287

9. Pearl R, Pancu D, Legome E. Hepatic abscess. J Emerg Med. 2005;28:337-339.doi:10.1016/j.jemermed.2004.08.024

10. Huang CJ, Pitt HA, Lipsett PA, et al. Pyogenic hepatic abscess. Changing trends over 42 years. Ann Surg. 1996;223(5):600-607; discussion 607-609.

11. Özgül E. Multiple pyogenic liver abscesses mimicking metastatic disease on computed tomography. Cureus. 2020;12(2):e7050. doi:10.7759/cureus.7050

12. Kuo SH, Lee YT, Li CR, et al. Mortality in Emergency Department Sepsis score as a prognostic indicator in patients with pyogenic liver abscess. Am J Emerg Med. 201331(6):916-921.

13. Lardière-Deguelte S, Ragot E, Amroun K, et al. Hepatic abscess: diagnosis and management. J Visc Surg. 2015;152(4):231-243. doi:10.1016/j.jviscsurg.2015.01.013

14. Halvorsen RA, Korobkin M, Foster WL, Silverman PM, Thompson WM. The variable CT appearance of hepatic abscesses. AJR Am J Roentgenol. 1984;142(5):941-946. doi:10.2214/ajr.142.5.941

15. Oh JG, Choi SY, Lee MH, et al. Differentiation of hepatic abscess from metastasis on contrast-enhanced dynamic computed tomography in patients with a history of extrahepatic malignancy: emphasis on dynamic change of arterial rim enhancement. Abdom Radiol (NY). 2019;44(2):529-538.

16. Jeffrey RB Jr, Tolentino CS, Chang FC, Federle MP. CT of small pyogenic hepatic abscesses: the cluster sign. AJR Am J Roentgenol. 1988;151(3):487-489. doi:10.2214/ajr.151.3.487

17. Mavilia MG, Molina M, Wu GY. The evolving nature of hepatic abscess: a review. J Clin Transl Hepatol. 2016;4(2):158-168. doi:10.14218/JCTH.2016.00004

Article PDF
Author and Disclosure Information

Jake A. Cresta, MDa; Michael A. Pavio, MDb; Jamie L. Lombardo, MDb; John G. McCarthy, MDa; Allison M. Bush, MDb

Correspondence:  Jake Cresta  (jcresta15@gmail.com)

aWalter Reed National Military Medical Center, Bethesda, Maryland

bNaval Medical Center Portsmouth, Virginia

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 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 informed consent for publication was obtained by the patient who was involved in this case.

Issue
Federal Practitioner - 41(5)s
Publications
Topics
Sections
Author and Disclosure Information

Jake A. Cresta, MDa; Michael A. Pavio, MDb; Jamie L. Lombardo, MDb; John G. McCarthy, MDa; Allison M. Bush, MDb

Correspondence:  Jake Cresta  (jcresta15@gmail.com)

aWalter Reed National Military Medical Center, Bethesda, Maryland

bNaval Medical Center Portsmouth, Virginia

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 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 informed consent for publication was obtained by the patient who was involved in this case.

Author and Disclosure Information

Jake A. Cresta, MDa; Michael A. Pavio, MDb; Jamie L. Lombardo, MDb; John G. McCarthy, MDa; Allison M. Bush, MDb

Correspondence:  Jake Cresta  (jcresta15@gmail.com)

aWalter Reed National Military Medical Center, Bethesda, Maryland

bNaval Medical Center Portsmouth, Virginia

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 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 informed consent for publication was obtained by the patient who was involved in this case.

Article PDF
Article PDF

Finding new liver lesions on imaging during a febrile illness may indicate hepatic abscesses or malignancy. These can be difficult to diagnose with imaging alone. Differentiating between infectious and neoplastic etiologies may require additional images and/or tissue samples.

Hepatic abscesses are more commonly seen with other abdominal or biliary infections while metastatic disease usually presents in patients with active cancer or on surveillance imaging. While renal cell carcinoma (RCC) is the most common malignant neoplasm of the kidney, chromophobe renal cell carcinoma (chRCC) is a rare subtype that comprises only 5% of RCC cases.1 We present a case of a patient with numerous new liver lesions and fever, initially thought to be hepatic abscesses, who was found to have metastatic chRCC.

 

CASE PRESENTATION

A 53 year-old male with a history of stage 2 chRCC and right radical nephrectomy 2 years prior presented to the emergency department following 1 week of drenching night sweats, fatigue, and subjective fevers. In addition, the patient reported gradually progressive, dull, right upper-quadrant abdominal pain. He noted no other acute medical complaints at the time of presentation. His history was notable for hyperlipidemia. His only surgery was the nephrectomy 2 years earlier. The patient reported no alcohol, tobacco, or drug use, any recent travel, or pet or animal exposure. On admission, he was afebrile with normal heart rate and was normotensive. His physical examination was remarkable for hepatomegaly with right upper-quadrant abdominal tenderness to palpation with a negative Murphy sign. There were otherwise no abnormal cardiovascular, respiratory, or skin findings.

Laboratory tests during initial evaluation were notable for hemoglobin of 10.0 g/dL, white blood cell count of 16.7×103 μL, alkaline phosphatase of 213 U/L, aspartate aminotransferase of 185 U/L, and alanine aminotransferase of 36 U/L. Screening tests for viral hepatitis A, B, and C were negative. Additional tests for HIV, rapid plasma reagin, Epstein-Barr virus, cytomegalovirus, and toxoplasma were negative. Tests for antimitochondrial, antismooth muscle, and serum antinuclear antibodies were negative.

figure_1.png

Chest X-ray did not reveal any acute cardiopulmonary process. Computed tomography with contrast of the abdomen and pelvis demonstrated numerous hypodensities within the right hepatic lobe. Right upper-quadrant ultrasound demonstrated multiple hyperechoic foci throughout the liver. confluent decreased T1 signal lesions with peripheral gadolinium hyperenhancement were evident on Gadolinium-enhanced T1-weighted magnetic resonance imaging (MRI) with fat saturation demonstrated numerous (Figure 1).

Liver biopsy and tissue cultures demonstrated normal hepatic tissue and no organism growth. Blood cultures demonstrated no growth. The patient was empirically treated with IV ceftriaxone 1 g daily and metronidazole 500 mg every 8 hours for suspected hepatic abscesses after he was admitted to the hospital.

figure_2.png

The patient’s symptoms initially improved following antibiotic treatment; however, he reported recurrence of the initial symptoms2 months later at a follow-up appointment with gastroenterology. Liver-associated enzymes also remained elevated despite 4 weeks of antibiotic treatment. Repeat gadolinium-enhanced T1 fat-saturated MRI demonstrated an interval increase in size and number of confluent hepatic lesions throughout the liver (Figure 2).

figure_4.png

figure_3.png

A repeat liver biopsy revealed metastatic chRCC (Figures 3 and 4) that was both morphologically and immunohistochemically similar to the first pathologic diagnosis made following nephrectomy. The first liver biopsy likely did not sample the metastatic lesions that were present but instead sampled the surrounding normal liver. The patient was initiated on lenvatinib and everolimus therapy with oncology, a recommended regimen per the National Comprehensive Cancer Network for patients with nonclear cell RCC.1

 

 

DISCUSSION

Chromophobe RCC is a rare form of RCC that has a recurrence-free survival of > 80% when treated in early stages.2 These neoplasms represent only 3000 to 6000 new cases of RCC annually, with an even lower incidence (6% to 7%) resulting in metastatic disease. The liver is the most common site of metastases (39%).2 Previously reported metastatic chRCC hepatic lesions have been incidentally noted on imaging with asymptomatic clinical presentations. In contrast to our patient, most documented cases report metastatic chRCC as a solitary hepatic lesion.3-7

A noteworthy genetic association with ChRCC is the Birt-Hogg-Dubé syndrome, which is an autosomal-dominant genetic disorder that results from germline mutations in the tumor suppressor folliculin gene located on chromosome 17.8 This syndrome is characterized by the development of various benign and malignant tumors, particularly chRCC. Our patient appeared to have a sporadic chRCC with the absence of other tumors and negative family history for malignancies. On his initial staging imaging, in accordance with National Comprehensive Cancer Network guidelines, our patient was identified only as having a 10-cm right renal mass and 1 benign regional lymph node with an otherwise unremarkable computed tomography of the chest, abdomen and pelvis, corresponding to stage 2 cancer.

Common causes of hepatic abscesses, the other potential diagnosis of concern for the patient, were biliary infections, portal vein ascension from abdominal sources, arterial translocation due to bacteremia, and local invasion due to suppuration of adjacent tissue.9 Incidence for hepatic abscesses increases with comorbidities such as diabetes, cirrhosis, malignancy, immunosuppression, and malnutrition.10Candida is also a common culprit when there are multiple, small abscesses, often in immunocompromised patients.11 Given the high mortality rates associated with hepatic abscesses, prompt treatment is imperative.10,12 Since the clinical signs and symptoms for hepatic abscesses are nonspecific (abdominal pain, fever, and malaise) and liver function tests can vary, the diagnosis primarily relies on imaging or tissue sampling.9

It can be difficult to distinguish abscesses from metastatic lesions based on imaging alone without microbiologic and pathologic confirmation.11,13,14 There are certain radiologic characteristics that have been found to favor abscesses over metastasis, including parenchymal enhancement, arterial rim enhancement, and perilesional hyperemia.15 However, previously described hallmark characteristics of hepatic abscesses, such as the “cluster sign” demonstrating early stages of abscess coalescence, have also been seen in some hepatic metastases.16

CONCLUSIONS

This patient highlights the presentation of a rare case of metastatic chRCC with multiple hepatic lesions. While often differentiated clinically, radiographically, or histologically, malignancy and abscess can be difficult to differentiate in a patient with fevers and leukocytosis with hepatic lesions.17 Early diagnosis of hepatic abscesses and initiation of antibiotic therapy are essential. In cases when it is challenging to characterize the hepatic lesions, repeated tissue sampling and imaging can help direct therapy. Attention should be paid to a previous history of malignancy and should raise suspicion for metastatic disease, particularly with misleading imaging studies and tissue samples.

Acknowledgments

This case was presented as a poster presentation at the Tri-Service American College of Physicians Meeting, September 7-10, 2022, San Antonio, Texas.

Finding new liver lesions on imaging during a febrile illness may indicate hepatic abscesses or malignancy. These can be difficult to diagnose with imaging alone. Differentiating between infectious and neoplastic etiologies may require additional images and/or tissue samples.

Hepatic abscesses are more commonly seen with other abdominal or biliary infections while metastatic disease usually presents in patients with active cancer or on surveillance imaging. While renal cell carcinoma (RCC) is the most common malignant neoplasm of the kidney, chromophobe renal cell carcinoma (chRCC) is a rare subtype that comprises only 5% of RCC cases.1 We present a case of a patient with numerous new liver lesions and fever, initially thought to be hepatic abscesses, who was found to have metastatic chRCC.

 

CASE PRESENTATION

A 53 year-old male with a history of stage 2 chRCC and right radical nephrectomy 2 years prior presented to the emergency department following 1 week of drenching night sweats, fatigue, and subjective fevers. In addition, the patient reported gradually progressive, dull, right upper-quadrant abdominal pain. He noted no other acute medical complaints at the time of presentation. His history was notable for hyperlipidemia. His only surgery was the nephrectomy 2 years earlier. The patient reported no alcohol, tobacco, or drug use, any recent travel, or pet or animal exposure. On admission, he was afebrile with normal heart rate and was normotensive. His physical examination was remarkable for hepatomegaly with right upper-quadrant abdominal tenderness to palpation with a negative Murphy sign. There were otherwise no abnormal cardiovascular, respiratory, or skin findings.

Laboratory tests during initial evaluation were notable for hemoglobin of 10.0 g/dL, white blood cell count of 16.7×103 μL, alkaline phosphatase of 213 U/L, aspartate aminotransferase of 185 U/L, and alanine aminotransferase of 36 U/L. Screening tests for viral hepatitis A, B, and C were negative. Additional tests for HIV, rapid plasma reagin, Epstein-Barr virus, cytomegalovirus, and toxoplasma were negative. Tests for antimitochondrial, antismooth muscle, and serum antinuclear antibodies were negative.

figure_1.png

Chest X-ray did not reveal any acute cardiopulmonary process. Computed tomography with contrast of the abdomen and pelvis demonstrated numerous hypodensities within the right hepatic lobe. Right upper-quadrant ultrasound demonstrated multiple hyperechoic foci throughout the liver. confluent decreased T1 signal lesions with peripheral gadolinium hyperenhancement were evident on Gadolinium-enhanced T1-weighted magnetic resonance imaging (MRI) with fat saturation demonstrated numerous (Figure 1).

Liver biopsy and tissue cultures demonstrated normal hepatic tissue and no organism growth. Blood cultures demonstrated no growth. The patient was empirically treated with IV ceftriaxone 1 g daily and metronidazole 500 mg every 8 hours for suspected hepatic abscesses after he was admitted to the hospital.

figure_2.png

The patient’s symptoms initially improved following antibiotic treatment; however, he reported recurrence of the initial symptoms2 months later at a follow-up appointment with gastroenterology. Liver-associated enzymes also remained elevated despite 4 weeks of antibiotic treatment. Repeat gadolinium-enhanced T1 fat-saturated MRI demonstrated an interval increase in size and number of confluent hepatic lesions throughout the liver (Figure 2).

figure_4.png

figure_3.png

A repeat liver biopsy revealed metastatic chRCC (Figures 3 and 4) that was both morphologically and immunohistochemically similar to the first pathologic diagnosis made following nephrectomy. The first liver biopsy likely did not sample the metastatic lesions that were present but instead sampled the surrounding normal liver. The patient was initiated on lenvatinib and everolimus therapy with oncology, a recommended regimen per the National Comprehensive Cancer Network for patients with nonclear cell RCC.1

 

 

DISCUSSION

Chromophobe RCC is a rare form of RCC that has a recurrence-free survival of > 80% when treated in early stages.2 These neoplasms represent only 3000 to 6000 new cases of RCC annually, with an even lower incidence (6% to 7%) resulting in metastatic disease. The liver is the most common site of metastases (39%).2 Previously reported metastatic chRCC hepatic lesions have been incidentally noted on imaging with asymptomatic clinical presentations. In contrast to our patient, most documented cases report metastatic chRCC as a solitary hepatic lesion.3-7

A noteworthy genetic association with ChRCC is the Birt-Hogg-Dubé syndrome, which is an autosomal-dominant genetic disorder that results from germline mutations in the tumor suppressor folliculin gene located on chromosome 17.8 This syndrome is characterized by the development of various benign and malignant tumors, particularly chRCC. Our patient appeared to have a sporadic chRCC with the absence of other tumors and negative family history for malignancies. On his initial staging imaging, in accordance with National Comprehensive Cancer Network guidelines, our patient was identified only as having a 10-cm right renal mass and 1 benign regional lymph node with an otherwise unremarkable computed tomography of the chest, abdomen and pelvis, corresponding to stage 2 cancer.

Common causes of hepatic abscesses, the other potential diagnosis of concern for the patient, were biliary infections, portal vein ascension from abdominal sources, arterial translocation due to bacteremia, and local invasion due to suppuration of adjacent tissue.9 Incidence for hepatic abscesses increases with comorbidities such as diabetes, cirrhosis, malignancy, immunosuppression, and malnutrition.10Candida is also a common culprit when there are multiple, small abscesses, often in immunocompromised patients.11 Given the high mortality rates associated with hepatic abscesses, prompt treatment is imperative.10,12 Since the clinical signs and symptoms for hepatic abscesses are nonspecific (abdominal pain, fever, and malaise) and liver function tests can vary, the diagnosis primarily relies on imaging or tissue sampling.9

It can be difficult to distinguish abscesses from metastatic lesions based on imaging alone without microbiologic and pathologic confirmation.11,13,14 There are certain radiologic characteristics that have been found to favor abscesses over metastasis, including parenchymal enhancement, arterial rim enhancement, and perilesional hyperemia.15 However, previously described hallmark characteristics of hepatic abscesses, such as the “cluster sign” demonstrating early stages of abscess coalescence, have also been seen in some hepatic metastases.16

CONCLUSIONS

This patient highlights the presentation of a rare case of metastatic chRCC with multiple hepatic lesions. While often differentiated clinically, radiographically, or histologically, malignancy and abscess can be difficult to differentiate in a patient with fevers and leukocytosis with hepatic lesions.17 Early diagnosis of hepatic abscesses and initiation of antibiotic therapy are essential. In cases when it is challenging to characterize the hepatic lesions, repeated tissue sampling and imaging can help direct therapy. Attention should be paid to a previous history of malignancy and should raise suspicion for metastatic disease, particularly with misleading imaging studies and tissue samples.

Acknowledgments

This case was presented as a poster presentation at the Tri-Service American College of Physicians Meeting, September 7-10, 2022, San Antonio, Texas.

References

1. National Comprehensive Cancer Network. Kidney cancer (version 2.2024). Accessed February 5, 2024. https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf

2. Vera-Badillo FE, Conde E, Duran I. Chromophobe renal cell carcinoma: a review of an uncommon entity. Int J Urol. 2012;19(10):894-900.doi:10.1111/j.1442-2042.2012.03079.x

3. Lordan JT, Fawcett WJ, Karanjia ND. Solitary liver metastasis of chromophobe renal cell carcinoma 20 years after nephrectomy treated by hepatic resection. Urology. 2008;72(1):230.e5-6. doi:10.1016/j.urology.2007.11.134

4. Talarico F, Buli P, Iusco D, Sangiorgi A, Jovine E. Synchronous nephrectomy and right hepatectomy for metastatic chromophobe renal cell carcinoma: report of a case and review of the literature. Chir Ital. 2007;59(2):257-261.

5. Aslam MI, Spencer L, Garcea G, et al. A case of liver metastasis from an oncocytoma with a focal area of chromophobe renal cell carcinoma: a wolf in sheep’s clothing. Int J Surg Pathol. 2008;17(2):158-162. doi:10.1177/1066896908318741

6. Kyoda Y, Kobayashi K, Takahashi A, et al. Liver metastasis with portal vein tumor thrombosis as a late recurrence of chromophobe renal cell carcinoma. Article in Japanese. Hinyokika Kiyo. 2009;55(1):23-25.

7. Talarico F, Capizzi D, Iusco DR. Solitary liver metastasis of chromophobe renal cell carcinoma 17 years after nephrectomy. a case report and review of the literature. Ann Ital Chir. 2013;84(ePub):S2239253X13021816.

8. Garje R, Elhag D, Yasin HA, Acharya L, Vaena D, Dahmoush L. Comprehensive review of chromophobe renal cell carcinoma. Crit Rev Oncol Hematol. 2021;160:103287. doi:10.1016/j.critrevonc.2021.103287

9. Pearl R, Pancu D, Legome E. Hepatic abscess. J Emerg Med. 2005;28:337-339.doi:10.1016/j.jemermed.2004.08.024

10. Huang CJ, Pitt HA, Lipsett PA, et al. Pyogenic hepatic abscess. Changing trends over 42 years. Ann Surg. 1996;223(5):600-607; discussion 607-609.

11. Özgül E. Multiple pyogenic liver abscesses mimicking metastatic disease on computed tomography. Cureus. 2020;12(2):e7050. doi:10.7759/cureus.7050

12. Kuo SH, Lee YT, Li CR, et al. Mortality in Emergency Department Sepsis score as a prognostic indicator in patients with pyogenic liver abscess. Am J Emerg Med. 201331(6):916-921.

13. Lardière-Deguelte S, Ragot E, Amroun K, et al. Hepatic abscess: diagnosis and management. J Visc Surg. 2015;152(4):231-243. doi:10.1016/j.jviscsurg.2015.01.013

14. Halvorsen RA, Korobkin M, Foster WL, Silverman PM, Thompson WM. The variable CT appearance of hepatic abscesses. AJR Am J Roentgenol. 1984;142(5):941-946. doi:10.2214/ajr.142.5.941

15. Oh JG, Choi SY, Lee MH, et al. Differentiation of hepatic abscess from metastasis on contrast-enhanced dynamic computed tomography in patients with a history of extrahepatic malignancy: emphasis on dynamic change of arterial rim enhancement. Abdom Radiol (NY). 2019;44(2):529-538.

16. Jeffrey RB Jr, Tolentino CS, Chang FC, Federle MP. CT of small pyogenic hepatic abscesses: the cluster sign. AJR Am J Roentgenol. 1988;151(3):487-489. doi:10.2214/ajr.151.3.487

17. Mavilia MG, Molina M, Wu GY. The evolving nature of hepatic abscess: a review. J Clin Transl Hepatol. 2016;4(2):158-168. doi:10.14218/JCTH.2016.00004

References

1. National Comprehensive Cancer Network. Kidney cancer (version 2.2024). Accessed February 5, 2024. https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf

2. Vera-Badillo FE, Conde E, Duran I. Chromophobe renal cell carcinoma: a review of an uncommon entity. Int J Urol. 2012;19(10):894-900.doi:10.1111/j.1442-2042.2012.03079.x

3. Lordan JT, Fawcett WJ, Karanjia ND. Solitary liver metastasis of chromophobe renal cell carcinoma 20 years after nephrectomy treated by hepatic resection. Urology. 2008;72(1):230.e5-6. doi:10.1016/j.urology.2007.11.134

4. Talarico F, Buli P, Iusco D, Sangiorgi A, Jovine E. Synchronous nephrectomy and right hepatectomy for metastatic chromophobe renal cell carcinoma: report of a case and review of the literature. Chir Ital. 2007;59(2):257-261.

5. Aslam MI, Spencer L, Garcea G, et al. A case of liver metastasis from an oncocytoma with a focal area of chromophobe renal cell carcinoma: a wolf in sheep’s clothing. Int J Surg Pathol. 2008;17(2):158-162. doi:10.1177/1066896908318741

6. Kyoda Y, Kobayashi K, Takahashi A, et al. Liver metastasis with portal vein tumor thrombosis as a late recurrence of chromophobe renal cell carcinoma. Article in Japanese. Hinyokika Kiyo. 2009;55(1):23-25.

7. Talarico F, Capizzi D, Iusco DR. Solitary liver metastasis of chromophobe renal cell carcinoma 17 years after nephrectomy. a case report and review of the literature. Ann Ital Chir. 2013;84(ePub):S2239253X13021816.

8. Garje R, Elhag D, Yasin HA, Acharya L, Vaena D, Dahmoush L. Comprehensive review of chromophobe renal cell carcinoma. Crit Rev Oncol Hematol. 2021;160:103287. doi:10.1016/j.critrevonc.2021.103287

9. Pearl R, Pancu D, Legome E. Hepatic abscess. J Emerg Med. 2005;28:337-339.doi:10.1016/j.jemermed.2004.08.024

10. Huang CJ, Pitt HA, Lipsett PA, et al. Pyogenic hepatic abscess. Changing trends over 42 years. Ann Surg. 1996;223(5):600-607; discussion 607-609.

11. Özgül E. Multiple pyogenic liver abscesses mimicking metastatic disease on computed tomography. Cureus. 2020;12(2):e7050. doi:10.7759/cureus.7050

12. Kuo SH, Lee YT, Li CR, et al. Mortality in Emergency Department Sepsis score as a prognostic indicator in patients with pyogenic liver abscess. Am J Emerg Med. 201331(6):916-921.

13. Lardière-Deguelte S, Ragot E, Amroun K, et al. Hepatic abscess: diagnosis and management. J Visc Surg. 2015;152(4):231-243. doi:10.1016/j.jviscsurg.2015.01.013

14. Halvorsen RA, Korobkin M, Foster WL, Silverman PM, Thompson WM. The variable CT appearance of hepatic abscesses. AJR Am J Roentgenol. 1984;142(5):941-946. doi:10.2214/ajr.142.5.941

15. Oh JG, Choi SY, Lee MH, et al. Differentiation of hepatic abscess from metastasis on contrast-enhanced dynamic computed tomography in patients with a history of extrahepatic malignancy: emphasis on dynamic change of arterial rim enhancement. Abdom Radiol (NY). 2019;44(2):529-538.

16. Jeffrey RB Jr, Tolentino CS, Chang FC, Federle MP. CT of small pyogenic hepatic abscesses: the cluster sign. AJR Am J Roentgenol. 1988;151(3):487-489. doi:10.2214/ajr.151.3.487

17. Mavilia MG, Molina M, Wu GY. The evolving nature of hepatic abscess: a review. J Clin Transl Hepatol. 2016;4(2):158-168. doi:10.14218/JCTH.2016.00004

Issue
Federal Practitioner - 41(5)s
Issue
Federal Practitioner - 41(5)s
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>0524 AVAHO RCC</fileName> <TBEID>0C02F265.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F265</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240308T112914</firstPublished> <LastPublished>20240428T220406</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240308T112914</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Jake A. Cresta, MDa; Michael A. Pavio, MDb; Jamie L. Lombardo, MDb; John G. McCarthy, MDa; Allison M. Bush, MDb</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>Finding new liver lesions on imaging during a febrile illness may indicate hepatic abscesses or malignancy. These can be difficult to diagnose with imaging alon</metaDescription> <articlePDF/> <teaserImage/> <title>A Case of Metastatic Chromophobe Renal Cell Carcinoma Masked as Suspected Hepatic Abscesses</title> <deck/> <eyebrow>Case in Point</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>2</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2953</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case in Point | 2953<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">45</term> </sections> <topics> <term canonical="true">263</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>A Case of Metastatic Chromophobe Renal Cell Carcinoma Masked as Suspected Hepatic Abscesses</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background: </b>Characterizing multiple hepatic lesions on cross-sectional imaging, particularly differentiating abscesses from metastatic lesions, can be challenging. <br/><br/><b>Case Presentation:</b> A male aged 53 years with a history of chromophobe renal cell carcinoma presented with fevers and abdominal pain and was found to have multiple hepatic lesions concerning for hepatic abscesses. The lesions initially evaded diagnosis on imaging, laboratory tests, and biopsy, but ultimately were determined to be a rare case of metastatic chromophobe renal cell carcinoma of the liver. <br/><br/><b>Conclusions:</b> The finding of multiple new liver lesions on imaging during a febrile illness is concerning for hepatic abscess or malignancy, which can be difficult to diagnose with imaging alone. Differentiation between infectious and neoplastic etiologies may require additional imaging and/or tissue sampling. </p> <p>Finding new liver lesions on imaging during a febrile illness may indicate hepatic abscesses or malignancy. These can be difficult to diagnose with imaging alone. Differentiating between infectious and neoplastic etiologies may require additional images and/or tissue samples.</p> <p>Hepatic abscesses are more commonly seen with other abdominal or biliary infections while metastatic disease usually presents in patients with active cancer or on surveillance imaging. While renal cell carcinoma (RCC) is the most common malignant neoplasm of the kidney, chromophobe renal cell carcinoma (chRCC) is a rare subtype that comprises only 5% of RCC cases.<sup>1</sup> We present a case of a patient with numerous new liver lesions and fever, initially thought to be hepatic abscesses, who was found to have metastatic chRCC.</p> <h2>CASE PRESENTATION</h2> <p>A 53 year-old male with a history of stage 2 chRCC and right radical nephrectomy 2 years prior presented to the emergency department following 1 week of drenching night sweats, fatigue, and subjective fevers. In addition, the patient reported gradually progressive, dull, right upper-quadrant abdominal pain. He noted no other acute medical complaints at the time of presentation. His history was notable for hyperlipidemia. His only surgery was the nephrectomy 2 years earlier. The patient reported no alcohol, tobacco, or drug use, any recent travel, or pet or animal exposure. On admission, he was afebrile with normal heart rate and was normotensive. His physical examination was remarkable for hepatomegaly with right upper-quadrant abdominal tenderness to palpation with a negative Murphy sign. There were otherwise no abnormal cardiovascular, respiratory, or skin findings.</p> <p>Laboratory tests during initial evaluation were notable for hemoglobin of 10.0 g/dL, white blood cell count of 16.7×10<sup>3</sup> μL, alkaline phosphatase of 213 U/L, aspartate aminotransferase of 185 U/L, and alanine aminotransferase of 36 U/L. Screening tests for viral hepatitis A, B, and C were negative. Additional tests for HIV, rapid plasma reagin, Epstein-Barr virus, cytomegalovirus, and toxoplasma were negative. Tests for antimitochondrial, antismooth muscle, and serum antinuclear antibodies were negative.<br/><br/>Chest X-ray did not reveal any acute cardiopulmonary process. Computed tomography with contrast of the abdomen and pelvis demonstrated numerous hypodensities within the right hepatic lobe. Right upper-quadrant ultrasound demonstrated multiple hyperechoic foci throughout the liver. confluent decreased T1 signal lesions with peripheral gadolinium hyperenhancement were evident on Gadolinium-enhanced T1-weighted magnetic resonance imaging (MRI) with fat saturation demonstrated numerous (Figure 1).<br/><br/>Liver biopsy and tissue cultures demonstrated normal hepatic tissue and no organism growth. Blood cultures demonstrated no growth. The patient was empirically treated with IV ceftriaxone 1 g daily and metronidazole 500 mg every 8 hours for suspected hepatic abscesses after he was admitted to the hospital.<br/><br/>The patient’s symptoms initially improved following antibiotic treatment; however, he reported recurrence of the initial symptoms2 months later at a follow-up appointment with gastroenterology. Liver-associated enzymes also remained elevated despite 4 weeks of antibiotic treatment. Repeat gadolinium-enhanced T1 fat-saturated MRI demonstrated an interval increase in size and number of confluent hepatic lesions throughout the liver (Figure 2).<br/><br/>A repeat liver biopsy revealed metastatic chRCC (Figures 3 and 4) that was both morphologically and immunohistochemically similar to the first pathologic diagnosis made following nephrectomy. The first liver biopsy likely did not sample the metastatic lesions that were present but instead sampled the surrounding normal liver. The patient was initiated on lenvatinib and everolimus therapy with oncology, a recommended regimen per the National Comprehensive Cancer Network for patients with nonclear cell RCC.<sup>1</sup></p> <h2>DISCUSSION</h2> <p>Chromophobe RCC is a rare form of RCC that has a recurrence-free survival of &gt; 80% when treated in early stages.<sup>2</sup> These neoplasms represent only 3000 to 6000 new cases of RCC annually, with an even lower incidence (6% to 7%) resulting in metastatic disease. The liver is the most common site of metastases (39%).<sup>2</sup> Previously reported metastatic chRCC hepatic lesions have been incidentally noted on imaging with asymptomatic clinical presentations. In contrast to our patient, most documented cases report metastatic chRCC as a solitary hepatic lesion.<sup>3-7</sup> </p> <p>A noteworthy genetic association with ChRCC is the Birt-Hogg-Dubé syndrome, which is an autosomal-dominant genetic disorder that results from germline mutations in the tumor suppressor folliculin gene located on chromosome 17.<sup>8</sup> This syndrome is characterized by the development of various benign and malignant tumors, particularly chRCC. Our patient appeared to have a sporadic chRCC with the absence of other tumors and negative family history for malignancies. On his initial staging imaging, in accordance with National Comprehensive Cancer Network guidelines, our patient was identified only as having a 10-cm right renal mass and 1 benign regional lymph node with an otherwise unremarkable computed tomography of the chest, abdomen and pelvis, corresponding to stage 2 cancer. <br/><br/>Common causes of hepatic abscesses, the other potential diagnosis of concern for the patient, were biliary infections, portal vein ascension from abdominal sources, arterial translocation due to bacteremia, and local invasion due to suppuration of adjacent tissue.<sup>9</sup> Incidence for hepatic abscesses increases with comorbidities such as diabetes, cirrhosis, malignancy, immunosuppression, and malnutrition.<sup>10</sup> <i>Candida</i> is also a common culprit when there are multiple, small abscesses, often in immunocompromised patients.<sup>11</sup> Given the high mortality rates associated with hepatic abscesses, prompt treatment is imperative.<sup>10,12</sup> Since the clinical signs and symptoms for hepatic abscesses are nonspecific (abdominal pain, fever, and malaise) and liver function tests can vary, the diagnosis primarily relies on imaging or tissue sampling.<sup>9</sup> <br/><br/>It can be difficult to distinguish abscesses from metastatic lesions based on imaging alone without microbiologic and pathologic confirmation.<sup>11,13,14</sup> There are certain radiologic characteristics that have been found to favor abscesses over metastasis, including parenchymal enhancement, arterial rim enhancement, and perilesional hyperemia.<sup>15</sup> However, previously described hallmark characteristics of hepatic abscesses, such as the “cluster sign” demonstrating early stages of abscess coalescence, have also been seen in some hepatic metastases.<sup>16</sup></p> <h2>CONCLUSIONS</h2> <p>This patient highlights the presentation of a rare case of metastatic chRCC with multiple hepatic lesions. While often differentiated clinically, radiographically, or histologically, malignancy and abscess can be difficult to differentiate in a patient with fevers and leukocytosis with hepatic lesions.<sup>17</sup> Early diagnosis of hepatic abscesses and initiation of antibiotic therapy are essential. In cases when it is challenging to characterize the hepatic lesions, repeated tissue sampling and imaging can help direct therapy. Attention should be paid to a previous history of malignancy and should raise suspicion for metastatic disease, particularly with misleading imaging studies and tissue samples. </p> <h3> Acknowledgments </h3> <p> <em>This case was presented as a poster presentation at the Tri-Service American College of Physicians Meeting, September 7-10, 2022, San Antonio, Texas.</em> </p> <h3> Author affiliations </h3> <p> <em><sup>a</sup>Walter Reed National Military Medical Center, Bethesda, Maryland<br/><br/><sup>b</sup>Naval Medical Center Portsmouth, Virginia</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 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 informed consent for publication was obtained by the patient who was involved in this case.</em> </p> <h3> References </h3> <p class="reference"> 1. National Comprehensive Cancer Network. Kidney cancer (version 2.2024). Accessed February 5, 2024. https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf<br/><br/> 2. Vera-Badillo FE, Conde E, Duran I. Chromophobe renal cell carcinoma: a review of an uncommon entity. <em>Int J Urol</em>. 2012;19(10):894-900.doi:10.1111/j.1442-2042.2012.03079.x<br/><br/> 3. Lordan JT, Fawcett WJ, Karanjia ND. Solitary liver metastasis of chromophobe renal cell carcinoma 20 years after nephrectomy treated by hepatic resection. <em>Urology</em>. 2008;72(1):230.e5-6. doi:10.1016/j.urology.2007.11.134<br/><br/> 4. Talarico F, Buli P, Iusco D, Sangiorgi A, Jovine E. Synchronous nephrectomy and right hepatectomy for metastatic chromophobe renal cell carcinoma: report of a case and review of the literature. <em>Chir Ital</em>. 2007;59(2):257-261.<br/><br/> 5. Aslam MI, Spencer L, Garcea G, et al. A case of liver metastasis from an oncocytoma with a focal area of chromophobe renal cell carcinoma: a wolf in sheep’s clothing. <em>Int J Surg Pathol</em>. 2008;17(2):158-162. doi:10.1177/1066896908318741<br/><br/> 6. Kyoda Y, Kobayashi K, Takahashi A, et al. Liver metastasis with portal vein tumor thrombosis as a late recurrence of chromophobe renal cell carcinoma. Article in Japanese. <em>Hinyokika Kiyo</em>. 2009;55(1):23-25.<br/><br/> 7. Talarico F, Capizzi D, Iusco DR. Solitary liver metastasis of chromophobe renal cell carcinoma 17 years after nephrectomy. a case report and review of the literature. <em>Ann Ital Chir</em>. 2013;84(ePub):S2239253X13021816.<br/><br/> 8. Garje R, Elhag D, Yasin HA, Acharya L, Vaena D, Dahmoush L. Comprehensive review of chromophobe renal cell carcinoma. <em>Crit Rev Oncol Hematol</em>. 2021;160:103287. doi:10.1016/j.critrevonc.2021.103287<br/><br/> 9. Pearl R, Pancu D, Legome E. Hepatic abscess. <em>J Emerg Med</em>. 2005;28:337-339.doi:10.1016/j.jemermed.2004.08.024<br/><br/>10. Huang CJ, Pitt HA, Lipsett PA, et al. Pyogenic hepatic abscess. Changing trends over 42 years. <em>Ann Surg</em>. 1996;223(5):600-607; discussion 607-609.<br/><br/>11. Özgül E. Multiple pyogenic liver abscesses mimicking metastatic disease on computed tomography. <em>Cureus</em>. 2020;12(2):e7050. doi:10.7759/cureus.7050<br/><br/>12. Kuo SH, Lee YT, Li CR, et al. Mortality in Emergency Department Sepsis score as a prognostic indicator in patients with pyogenic liver abscess. <em>Am J Emerg Med</em>. 201331(6):916-921.<br/><br/>13. Lardière-Deguelte S, Ragot E, Amroun K, et al. Hepatic abscess: diagnosis and management. <em>J Visc Surg</em>. 2015;152(4):231-243. doi:10.1016/j.jviscsurg.2015.01.013 <br/><br/>14. Halvorsen RA, Korobkin M, Foster WL, Silverman PM, Thompson WM. The variable CT appearance of hepatic abscesses. <em>AJR Am J Roentgenol</em>. 1984;142(5):941-946. doi:10.2214/ajr.142.5.941<br/><br/>15. Oh JG, Choi SY, Lee MH, et al. Differentiation of hepatic abscess from metastasis on contrast-enhanced dynamic computed tomography in patients with a history of extrahepatic malignancy: emphasis on dynamic change of arterial rim enhancement. <em>Abdom Radiol (NY)</em>. 2019;44(2):529-538.<br/><br/>16. Jeffrey RB Jr, Tolentino CS, Chang FC, Federle MP. CT of small pyogenic hepatic abscesses: the cluster sign. <em>AJR Am J Roentgenol</em>. 1988;151(3):487-489. doi:10.2214/ajr.151.3.487<br/><br/>17. Mavilia MG, Molina M, Wu GY. The evolving nature of hepatic abscess: a review. <em>J Clin Transl Hepatol</em>. 2016;4(2):158-168. doi:10.14218/JCTH.2016.00004</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

Medication-Nonadherent Hypothyroidism Requiring Frequent Primary Care Visits to Achieve Euthyroidism

Article Type
Changed
Thu, 03/07/2024 - 12:47

Nonadherence to medications is an issue across health care. In endocrinology, hypothyroidism, a deficiency of thyroid hormones, is most often treated with levothyroxine and if left untreated can lead to myxedema coma, which can lead to death due to multiorgan dysfunction.1 Therefore, adherence to levothyroxine is very important in preventing fatal complications.

We present the case of a patient with persistent primary hypothyroidism who was suspected to be nonadherent to levothyroxine, although the patient consistently claimed adherence. The patient’s plasma thyrotropin (TSH) level improved to reference range after 6 weeks of weekly primary care clinic visits. After stopping the visits, his plasma TSH level increased again, so 9 more weeks of visits resumed, which again helped bring down his plasma TSH levels.

Case Presentation

A male patient aged 67 years presented to the Dayton Veterans Affairs Medical Center (VAMC) endocrinology clinic for evaluation of thyroid nodules. The patient reported no history of neck irradiation and a physical examination was unremarkable. At that time, laboratory results showed a slightly elevated plasma TSH level of 4.35 uIU/mL (reference range, 0.35-4.00 uIU/mL) and normal free thyroxine (T4) of 1.00 ng/dL (reference range, 0.74-1.46 ng/dL). Later that year, the patient underwent a total thyroidectomy at the Cincinnati VAMC for Hurthle cell variant papillary thyroid carcinoma that was noted on biopsy at the Dayton VAMC. After surgical pathology results were available, the patient started levothyroxine 200 mcg daily, although 224 mcg would have been more appropriate based on his 142 kg weight. Due to a history of arrhythmia, the goal plasma TSH level was 0.10 to 0.50 uIU/mL. The patient subsequently underwent radioactive iodine ablation. After levothyroxine dose adjustments, the patient’s plasma TSH level was noted to be within his target range at 0.28 uIU/mL 3 months postablation.

Over the next 5 years the patient had regular laboratory tests during which his plasma TSH level rose and were typically high despite adjusting levothyroxine doses between 200 mcg and 325 mcg. The patient received counseling on taking the medication in the morning on an empty stomach and waiting at least 1 hour before consuming anything, and he went to many follow-up visits at the Dayton VAMC endocrinology clinic. He reported no vomiting or diarrhea but endorsed weight gain once. The patient also had high free T4 at times and did not take extra levothyroxine before undergoing laboratory tests.

Nonadherence to levothyroxine was suspected, but the patient insisted he was adherent. He received the medication in the mail regularly, generally had 90-day refills unless a dose change was made, used a pill box, and had social support from his son, but he did not use a phone alarm to remind him to take it. A home care nurse made weekly visits to make sure the remaining levothyroxine pill counts were correct; however, the patient continued to have difficulty maintaining daily adherence at home as indicated by the nurse’s pill counts not aligning with the number of pills which should have been left if the patient was talking the pills daily.

table.png

The patient was asked to visit a local community-based outpatient clinic (CBOC) weekly (to avoid patient travel time to Dayton VAMC > 1 hour) to check pill counts and assess adherence. The patient went to the CBOC clinic for these visits, during which pill counts indicated much better but not 100% adherence. After 6 weeks of clinic visits, his plasma TSH decreased to 1.01 uIU/mL, which was within the reference range, and the patient stopped coming to the weekly clinic visits (Table). Four months later, the patient's plasma TSH levels increased to 80.72 uIU/mL. Nonadherence to levothyroxine was suspected again. He was asked to resume weekly clinic visits, and the life-threatening effects of hypothyroidism and not taking levothyroxine were discussed with the patient and his son. The patient made CBOC clinic visits for 9 weeks, after which his plasma TSH level was low at 0.23 uIU/mL.

 

 

Discussion

There are multiple important causes to consider in patients with persistent hypothyroidism. One is medication nonadherence, which was most likely seen in the patient in this case. Missing even 1 day of levothyroxine can affect TSH and thyroid hormone levels for several days due to the long half-life of the medication.2 Hepp and colleagues found that patients with hypothyroidism were significantly more likely to be nonadherent to levothyroxine if they had comorbid conditions such as type 2 diabetes or were obese.3 Another study of levothyroxine adherence found that the most common reason for missing doses was forgetfulness.4 However, memory and cognition impairments can also be symptoms of hypothyroidism itself; Haskard-Zolnierek and colleagues found a significant association between nonadherence to levothyroxine and self-reported brain fog in patients with hypothyroidism.5

Another cause of persistent hypothyroidism is malabsorption. Absorption of levothyroxine can be affected by intestinal malabsorption due to inflammatory bowel disease, lactose intolerance, or gastrointestinal infection, as well as several foods, drinks (eg, coffee), medications, vitamins, and supplements (eg, proton-pump inhibitors and calcium).2,6 Levothyroxine is absorbed mainly at the jejunum and upper ileum, so any pathologies or ingested items that would directly or indirectly affect absorption at those sites can affect levothyroxine absorption.2

A liquid levothyroxine formulation can help with malabsorption.2 Alternatively, weight gain may lead to a need for increasing the dosage of levothyroxine.2,6 Other factors that can affect TSH levels include Addison disease, dysregulation of the hypothalamic-pituitary-thyroid axis, and TSH heterophile antibodies.2

Research describes methods that have effectively treated hypothyroidism in patients struggling with levothyroxine adherence. Two case reports describe weekly visits for levothyroxine administration successfully treating uncontrolled hypothyroidism.7,8 A meta-analysis found that while weekly levothyroxine tablets led to a higher mean TSH level than daily use, weekly use still led to reference-range TSH levels, suggesting that weekly levothyroxine may be a helpful alternative for nonadherent patients.9 Alternatively, patients taking levothyroxine tablets have been shown to forget to take their medication more frequently compared to those taking the liquid formulation.10,11 Additionally, a study by El Helou and colleagues found that adherence to levothyroxine was significantly improved when patients had endocrinology visits once a month and when the endocrinologist provided information about hypothyroidism.12

Another method that may improve adherence to levothyroxine is telehealth visits. This would be especially helpful for patients who live far from the clinic or do not have the time, transportation, or financial means to visit the clinic for weekly visits to assess medication adherence. Additionally, patients may be afraid of admitting to a health care professional that they are nonadherent. Clinicians must be tactful when asking about adherence to make the patient feel comfortable with admitting to nonadherence if their cognition is not impaired. Then, a patient-led conversation can occur regarding realistic ways the patient feels they can work toward adherence.

To our knowledge, the patient in this case report had no symptoms of intestinal malabsorption, and weight gain was not thought to be the issue, as levothyroxine dosage was adjusted multiple times. His plasma TSH levels returned to reference range after weekly pill count visits for 6 weeks and after weekly pill count visits for 9 weeks. Therefore, nonadherence to levothyroxine was suspected to be the cause of frequently elevated plasma TSH levels despite the patient’s insistence on adherence. While the patient did not report memory issues, cognitive impairments due to hypothyroidism may have been contributing to his probable nonadherence. Additionally, he had comorbidities, such as type 2 diabetes mellitus and obesity, which may have made adherence more difficult.

Levothyroxine was also only prescribed in daily tablet form, so the frequency and formulation may have also contributed to nonadherence. While the home nurse was originally sent to assess the patient’s adherence, the care team could have had the nurse start giving the patient weekly levothyroxine once nonadherence was determined to be a likely issue. The patient’s adherence only improved when he went to the clinic for pill counts but not when the home nurse came to his house weekly; this could be because the patient knew he had to invest the time to physically go to clinic visits for pill checks, motivating him to increase adherence.

Conclusions

This case reports a patient with frequently high plasma TSH levels achieving normalization of plasma TSH levels after weekly medication adherence checks at a primary care clinic. Weekly visits to a clinic seem impractical compared to weekly dosing with a visiting nurse; however, after review of the literature, this may be an approach to consider in the future. This strategy may especially help in cases of persistent abnormal plasma TSH levels in which no etiology can be found other than suspected medication nonadherence. Knowing their medication use will be checked at weekly clinic visits may motivate patients to be adherent.

References

1. Chaker L, Bianco AC, Jonklaas J, Peeters RP. Hypothyroidism. Lancet. 2017;390(10101):1550-1562. doi:10.1016/S0140-6736(17)30703-1

2. Centanni M, Benvenga S, Sachmechi I. Diagnosis and management of treatment-refractory hypothyroidism: an expert consensus report. J Endocrinol Invest. 2017;40(12):1289-1301. doi:10.1007/s40618-017-0706-y

3. Hepp Z, Lage MJ, Espaillat R, Gossain VV. The association between adherence to levothyroxine and economic and clinical outcomes in patients with hypothyroidism in the US. J Med Econ. 2018;21(9):912-919. doi:10.1080/13696998.2018.1484749

4. Shakya Shrestha S, Risal K, Shrestha R, Bhatta RD. Medication Adherence to Levothyroxine Therapy among Hypothyroid Patients and their Clinical Outcomes with Special Reference to Thyroid Function Parameters. Kathmandu Univ Med J (KUMJ). 2018;16(62):129-137.

5. Haskard-Zolnierek K, Wilson C, Pruin J, Deason R, Howard K. The Relationship Between Brain Fog and Medication Adherence for Individuals With Hypothyroidism. Clin Nurs Res. 2022;31(3):445-452. doi:10.1177/10547738211038127

6. McNally LJ, Ofiaeli CI, Oyibo SO. Treatment-refractory hypothyroidism. BMJ. 2019;364:l579. Published 2019 Feb 25. doi:10.1136/bmj.l579

7. Nakano Y, Hashimoto K, Ohkiba N, et al. A Case of Refractory Hypothyroidism due to Poor Compliance Treated with the Weekly Intravenous and Oral Levothyroxine Administration. Case Rep Endocrinol. 2019;2019:5986014. Published 2019 Feb 5. doi:10.1155/2019/5986014

8. Kiran Z, Shaikh KS, Fatima N, Tariq N, Baloch AA. Levothyroxine absorption test followed by directly observed treatment on an outpatient basis to address long-term high TSH levels in a hypothyroid patient: a case report. J Med Case Rep. 2023;17(1):24. Published 2023 Jan 25. doi:10.1186/s13256-023-03760-0

9. Chiu HH, Larrazabal R Jr, Uy AB, Jimeno C. Weekly Versus Daily Levothyroxine Tablet Replacement in Adults with Hypothyroidism: A Meta-Analysis. J ASEAN Fed Endocr Soc. 2021;36(2):156-160. doi:10.15605/jafes.036.02.07

10. Cappelli C, Castello R, Marini F, et al. Adherence to Levothyroxine Treatment Among Patients With Hypothyroidism: A Northeastern Italian Survey. Front Endocrinol (Lausanne). 2018;9:699. Published 2018 Nov 23. doi:10.3389/fendo.2018.00699

11. Bocale R, Desideri G, Barini A, et al. Long-Term Adherence to Levothyroxine Replacement Therapy in Thyroidectomized Patients. J Clin Med. 2022;11(15):4296. Published 2022 Jul 24. doi:10.3390/jcm11154296

12. El Helou S, Hallit S, Awada S, et al. Adherence to levothyroxine among patients with hypothyroidism in Lebanon. East Mediterr Health J. 2019;25(3):149-159. Published 2019 Apr 25. doi:10.26719/emhj.18.022

Article PDF
Author and Disclosure Information

Sabrina Kaul;a,b Ankur Gupta, MDa,b

Correspondence: Ankur Gupta (ankur.gupta@va.gov)

aDayton Veterans Affairs Medical Center, Ohio

bWright State University Boonshoft School of Medicine, Dayton, Ohio

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 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 drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

Written informed consent was obtained from the patient presented in this case report. Patient identifiers have been removed to protect the privacy of the patient.

Issue
Federal Practitioner - 41(3)a
Publications
Topics
Page Number
84
Sections
Author and Disclosure Information

Sabrina Kaul;a,b Ankur Gupta, MDa,b

Correspondence: Ankur Gupta (ankur.gupta@va.gov)

aDayton Veterans Affairs Medical Center, Ohio

bWright State University Boonshoft School of Medicine, Dayton, Ohio

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 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 drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

Written informed consent was obtained from the patient presented in this case report. Patient identifiers have been removed to protect the privacy of the patient.

Author and Disclosure Information

Sabrina Kaul;a,b Ankur Gupta, MDa,b

Correspondence: Ankur Gupta (ankur.gupta@va.gov)

aDayton Veterans Affairs Medical Center, Ohio

bWright State University Boonshoft School of Medicine, Dayton, Ohio

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 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 drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

Written informed consent was obtained from the patient presented in this case report. Patient identifiers have been removed to protect the privacy of the patient.

Article PDF
Article PDF

Nonadherence to medications is an issue across health care. In endocrinology, hypothyroidism, a deficiency of thyroid hormones, is most often treated with levothyroxine and if left untreated can lead to myxedema coma, which can lead to death due to multiorgan dysfunction.1 Therefore, adherence to levothyroxine is very important in preventing fatal complications.

We present the case of a patient with persistent primary hypothyroidism who was suspected to be nonadherent to levothyroxine, although the patient consistently claimed adherence. The patient’s plasma thyrotropin (TSH) level improved to reference range after 6 weeks of weekly primary care clinic visits. After stopping the visits, his plasma TSH level increased again, so 9 more weeks of visits resumed, which again helped bring down his plasma TSH levels.

Case Presentation

A male patient aged 67 years presented to the Dayton Veterans Affairs Medical Center (VAMC) endocrinology clinic for evaluation of thyroid nodules. The patient reported no history of neck irradiation and a physical examination was unremarkable. At that time, laboratory results showed a slightly elevated plasma TSH level of 4.35 uIU/mL (reference range, 0.35-4.00 uIU/mL) and normal free thyroxine (T4) of 1.00 ng/dL (reference range, 0.74-1.46 ng/dL). Later that year, the patient underwent a total thyroidectomy at the Cincinnati VAMC for Hurthle cell variant papillary thyroid carcinoma that was noted on biopsy at the Dayton VAMC. After surgical pathology results were available, the patient started levothyroxine 200 mcg daily, although 224 mcg would have been more appropriate based on his 142 kg weight. Due to a history of arrhythmia, the goal plasma TSH level was 0.10 to 0.50 uIU/mL. The patient subsequently underwent radioactive iodine ablation. After levothyroxine dose adjustments, the patient’s plasma TSH level was noted to be within his target range at 0.28 uIU/mL 3 months postablation.

Over the next 5 years the patient had regular laboratory tests during which his plasma TSH level rose and were typically high despite adjusting levothyroxine doses between 200 mcg and 325 mcg. The patient received counseling on taking the medication in the morning on an empty stomach and waiting at least 1 hour before consuming anything, and he went to many follow-up visits at the Dayton VAMC endocrinology clinic. He reported no vomiting or diarrhea but endorsed weight gain once. The patient also had high free T4 at times and did not take extra levothyroxine before undergoing laboratory tests.

Nonadherence to levothyroxine was suspected, but the patient insisted he was adherent. He received the medication in the mail regularly, generally had 90-day refills unless a dose change was made, used a pill box, and had social support from his son, but he did not use a phone alarm to remind him to take it. A home care nurse made weekly visits to make sure the remaining levothyroxine pill counts were correct; however, the patient continued to have difficulty maintaining daily adherence at home as indicated by the nurse’s pill counts not aligning with the number of pills which should have been left if the patient was talking the pills daily.

table.png

The patient was asked to visit a local community-based outpatient clinic (CBOC) weekly (to avoid patient travel time to Dayton VAMC > 1 hour) to check pill counts and assess adherence. The patient went to the CBOC clinic for these visits, during which pill counts indicated much better but not 100% adherence. After 6 weeks of clinic visits, his plasma TSH decreased to 1.01 uIU/mL, which was within the reference range, and the patient stopped coming to the weekly clinic visits (Table). Four months later, the patient's plasma TSH levels increased to 80.72 uIU/mL. Nonadherence to levothyroxine was suspected again. He was asked to resume weekly clinic visits, and the life-threatening effects of hypothyroidism and not taking levothyroxine were discussed with the patient and his son. The patient made CBOC clinic visits for 9 weeks, after which his plasma TSH level was low at 0.23 uIU/mL.

 

 

Discussion

There are multiple important causes to consider in patients with persistent hypothyroidism. One is medication nonadherence, which was most likely seen in the patient in this case. Missing even 1 day of levothyroxine can affect TSH and thyroid hormone levels for several days due to the long half-life of the medication.2 Hepp and colleagues found that patients with hypothyroidism were significantly more likely to be nonadherent to levothyroxine if they had comorbid conditions such as type 2 diabetes or were obese.3 Another study of levothyroxine adherence found that the most common reason for missing doses was forgetfulness.4 However, memory and cognition impairments can also be symptoms of hypothyroidism itself; Haskard-Zolnierek and colleagues found a significant association between nonadherence to levothyroxine and self-reported brain fog in patients with hypothyroidism.5

Another cause of persistent hypothyroidism is malabsorption. Absorption of levothyroxine can be affected by intestinal malabsorption due to inflammatory bowel disease, lactose intolerance, or gastrointestinal infection, as well as several foods, drinks (eg, coffee), medications, vitamins, and supplements (eg, proton-pump inhibitors and calcium).2,6 Levothyroxine is absorbed mainly at the jejunum and upper ileum, so any pathologies or ingested items that would directly or indirectly affect absorption at those sites can affect levothyroxine absorption.2

A liquid levothyroxine formulation can help with malabsorption.2 Alternatively, weight gain may lead to a need for increasing the dosage of levothyroxine.2,6 Other factors that can affect TSH levels include Addison disease, dysregulation of the hypothalamic-pituitary-thyroid axis, and TSH heterophile antibodies.2

Research describes methods that have effectively treated hypothyroidism in patients struggling with levothyroxine adherence. Two case reports describe weekly visits for levothyroxine administration successfully treating uncontrolled hypothyroidism.7,8 A meta-analysis found that while weekly levothyroxine tablets led to a higher mean TSH level than daily use, weekly use still led to reference-range TSH levels, suggesting that weekly levothyroxine may be a helpful alternative for nonadherent patients.9 Alternatively, patients taking levothyroxine tablets have been shown to forget to take their medication more frequently compared to those taking the liquid formulation.10,11 Additionally, a study by El Helou and colleagues found that adherence to levothyroxine was significantly improved when patients had endocrinology visits once a month and when the endocrinologist provided information about hypothyroidism.12

Another method that may improve adherence to levothyroxine is telehealth visits. This would be especially helpful for patients who live far from the clinic or do not have the time, transportation, or financial means to visit the clinic for weekly visits to assess medication adherence. Additionally, patients may be afraid of admitting to a health care professional that they are nonadherent. Clinicians must be tactful when asking about adherence to make the patient feel comfortable with admitting to nonadherence if their cognition is not impaired. Then, a patient-led conversation can occur regarding realistic ways the patient feels they can work toward adherence.

To our knowledge, the patient in this case report had no symptoms of intestinal malabsorption, and weight gain was not thought to be the issue, as levothyroxine dosage was adjusted multiple times. His plasma TSH levels returned to reference range after weekly pill count visits for 6 weeks and after weekly pill count visits for 9 weeks. Therefore, nonadherence to levothyroxine was suspected to be the cause of frequently elevated plasma TSH levels despite the patient’s insistence on adherence. While the patient did not report memory issues, cognitive impairments due to hypothyroidism may have been contributing to his probable nonadherence. Additionally, he had comorbidities, such as type 2 diabetes mellitus and obesity, which may have made adherence more difficult.

Levothyroxine was also only prescribed in daily tablet form, so the frequency and formulation may have also contributed to nonadherence. While the home nurse was originally sent to assess the patient’s adherence, the care team could have had the nurse start giving the patient weekly levothyroxine once nonadherence was determined to be a likely issue. The patient’s adherence only improved when he went to the clinic for pill counts but not when the home nurse came to his house weekly; this could be because the patient knew he had to invest the time to physically go to clinic visits for pill checks, motivating him to increase adherence.

Conclusions

This case reports a patient with frequently high plasma TSH levels achieving normalization of plasma TSH levels after weekly medication adherence checks at a primary care clinic. Weekly visits to a clinic seem impractical compared to weekly dosing with a visiting nurse; however, after review of the literature, this may be an approach to consider in the future. This strategy may especially help in cases of persistent abnormal plasma TSH levels in which no etiology can be found other than suspected medication nonadherence. Knowing their medication use will be checked at weekly clinic visits may motivate patients to be adherent.

Nonadherence to medications is an issue across health care. In endocrinology, hypothyroidism, a deficiency of thyroid hormones, is most often treated with levothyroxine and if left untreated can lead to myxedema coma, which can lead to death due to multiorgan dysfunction.1 Therefore, adherence to levothyroxine is very important in preventing fatal complications.

We present the case of a patient with persistent primary hypothyroidism who was suspected to be nonadherent to levothyroxine, although the patient consistently claimed adherence. The patient’s plasma thyrotropin (TSH) level improved to reference range after 6 weeks of weekly primary care clinic visits. After stopping the visits, his plasma TSH level increased again, so 9 more weeks of visits resumed, which again helped bring down his plasma TSH levels.

Case Presentation

A male patient aged 67 years presented to the Dayton Veterans Affairs Medical Center (VAMC) endocrinology clinic for evaluation of thyroid nodules. The patient reported no history of neck irradiation and a physical examination was unremarkable. At that time, laboratory results showed a slightly elevated plasma TSH level of 4.35 uIU/mL (reference range, 0.35-4.00 uIU/mL) and normal free thyroxine (T4) of 1.00 ng/dL (reference range, 0.74-1.46 ng/dL). Later that year, the patient underwent a total thyroidectomy at the Cincinnati VAMC for Hurthle cell variant papillary thyroid carcinoma that was noted on biopsy at the Dayton VAMC. After surgical pathology results were available, the patient started levothyroxine 200 mcg daily, although 224 mcg would have been more appropriate based on his 142 kg weight. Due to a history of arrhythmia, the goal plasma TSH level was 0.10 to 0.50 uIU/mL. The patient subsequently underwent radioactive iodine ablation. After levothyroxine dose adjustments, the patient’s plasma TSH level was noted to be within his target range at 0.28 uIU/mL 3 months postablation.

Over the next 5 years the patient had regular laboratory tests during which his plasma TSH level rose and were typically high despite adjusting levothyroxine doses between 200 mcg and 325 mcg. The patient received counseling on taking the medication in the morning on an empty stomach and waiting at least 1 hour before consuming anything, and he went to many follow-up visits at the Dayton VAMC endocrinology clinic. He reported no vomiting or diarrhea but endorsed weight gain once. The patient also had high free T4 at times and did not take extra levothyroxine before undergoing laboratory tests.

Nonadherence to levothyroxine was suspected, but the patient insisted he was adherent. He received the medication in the mail regularly, generally had 90-day refills unless a dose change was made, used a pill box, and had social support from his son, but he did not use a phone alarm to remind him to take it. A home care nurse made weekly visits to make sure the remaining levothyroxine pill counts were correct; however, the patient continued to have difficulty maintaining daily adherence at home as indicated by the nurse’s pill counts not aligning with the number of pills which should have been left if the patient was talking the pills daily.

table.png

The patient was asked to visit a local community-based outpatient clinic (CBOC) weekly (to avoid patient travel time to Dayton VAMC > 1 hour) to check pill counts and assess adherence. The patient went to the CBOC clinic for these visits, during which pill counts indicated much better but not 100% adherence. After 6 weeks of clinic visits, his plasma TSH decreased to 1.01 uIU/mL, which was within the reference range, and the patient stopped coming to the weekly clinic visits (Table). Four months later, the patient's plasma TSH levels increased to 80.72 uIU/mL. Nonadherence to levothyroxine was suspected again. He was asked to resume weekly clinic visits, and the life-threatening effects of hypothyroidism and not taking levothyroxine were discussed with the patient and his son. The patient made CBOC clinic visits for 9 weeks, after which his plasma TSH level was low at 0.23 uIU/mL.

 

 

Discussion

There are multiple important causes to consider in patients with persistent hypothyroidism. One is medication nonadherence, which was most likely seen in the patient in this case. Missing even 1 day of levothyroxine can affect TSH and thyroid hormone levels for several days due to the long half-life of the medication.2 Hepp and colleagues found that patients with hypothyroidism were significantly more likely to be nonadherent to levothyroxine if they had comorbid conditions such as type 2 diabetes or were obese.3 Another study of levothyroxine adherence found that the most common reason for missing doses was forgetfulness.4 However, memory and cognition impairments can also be symptoms of hypothyroidism itself; Haskard-Zolnierek and colleagues found a significant association between nonadherence to levothyroxine and self-reported brain fog in patients with hypothyroidism.5

Another cause of persistent hypothyroidism is malabsorption. Absorption of levothyroxine can be affected by intestinal malabsorption due to inflammatory bowel disease, lactose intolerance, or gastrointestinal infection, as well as several foods, drinks (eg, coffee), medications, vitamins, and supplements (eg, proton-pump inhibitors and calcium).2,6 Levothyroxine is absorbed mainly at the jejunum and upper ileum, so any pathologies or ingested items that would directly or indirectly affect absorption at those sites can affect levothyroxine absorption.2

A liquid levothyroxine formulation can help with malabsorption.2 Alternatively, weight gain may lead to a need for increasing the dosage of levothyroxine.2,6 Other factors that can affect TSH levels include Addison disease, dysregulation of the hypothalamic-pituitary-thyroid axis, and TSH heterophile antibodies.2

Research describes methods that have effectively treated hypothyroidism in patients struggling with levothyroxine adherence. Two case reports describe weekly visits for levothyroxine administration successfully treating uncontrolled hypothyroidism.7,8 A meta-analysis found that while weekly levothyroxine tablets led to a higher mean TSH level than daily use, weekly use still led to reference-range TSH levels, suggesting that weekly levothyroxine may be a helpful alternative for nonadherent patients.9 Alternatively, patients taking levothyroxine tablets have been shown to forget to take their medication more frequently compared to those taking the liquid formulation.10,11 Additionally, a study by El Helou and colleagues found that adherence to levothyroxine was significantly improved when patients had endocrinology visits once a month and when the endocrinologist provided information about hypothyroidism.12

Another method that may improve adherence to levothyroxine is telehealth visits. This would be especially helpful for patients who live far from the clinic or do not have the time, transportation, or financial means to visit the clinic for weekly visits to assess medication adherence. Additionally, patients may be afraid of admitting to a health care professional that they are nonadherent. Clinicians must be tactful when asking about adherence to make the patient feel comfortable with admitting to nonadherence if their cognition is not impaired. Then, a patient-led conversation can occur regarding realistic ways the patient feels they can work toward adherence.

To our knowledge, the patient in this case report had no symptoms of intestinal malabsorption, and weight gain was not thought to be the issue, as levothyroxine dosage was adjusted multiple times. His plasma TSH levels returned to reference range after weekly pill count visits for 6 weeks and after weekly pill count visits for 9 weeks. Therefore, nonadherence to levothyroxine was suspected to be the cause of frequently elevated plasma TSH levels despite the patient’s insistence on adherence. While the patient did not report memory issues, cognitive impairments due to hypothyroidism may have been contributing to his probable nonadherence. Additionally, he had comorbidities, such as type 2 diabetes mellitus and obesity, which may have made adherence more difficult.

Levothyroxine was also only prescribed in daily tablet form, so the frequency and formulation may have also contributed to nonadherence. While the home nurse was originally sent to assess the patient’s adherence, the care team could have had the nurse start giving the patient weekly levothyroxine once nonadherence was determined to be a likely issue. The patient’s adherence only improved when he went to the clinic for pill counts but not when the home nurse came to his house weekly; this could be because the patient knew he had to invest the time to physically go to clinic visits for pill checks, motivating him to increase adherence.

Conclusions

This case reports a patient with frequently high plasma TSH levels achieving normalization of plasma TSH levels after weekly medication adherence checks at a primary care clinic. Weekly visits to a clinic seem impractical compared to weekly dosing with a visiting nurse; however, after review of the literature, this may be an approach to consider in the future. This strategy may especially help in cases of persistent abnormal plasma TSH levels in which no etiology can be found other than suspected medication nonadherence. Knowing their medication use will be checked at weekly clinic visits may motivate patients to be adherent.

References

1. Chaker L, Bianco AC, Jonklaas J, Peeters RP. Hypothyroidism. Lancet. 2017;390(10101):1550-1562. doi:10.1016/S0140-6736(17)30703-1

2. Centanni M, Benvenga S, Sachmechi I. Diagnosis and management of treatment-refractory hypothyroidism: an expert consensus report. J Endocrinol Invest. 2017;40(12):1289-1301. doi:10.1007/s40618-017-0706-y

3. Hepp Z, Lage MJ, Espaillat R, Gossain VV. The association between adherence to levothyroxine and economic and clinical outcomes in patients with hypothyroidism in the US. J Med Econ. 2018;21(9):912-919. doi:10.1080/13696998.2018.1484749

4. Shakya Shrestha S, Risal K, Shrestha R, Bhatta RD. Medication Adherence to Levothyroxine Therapy among Hypothyroid Patients and their Clinical Outcomes with Special Reference to Thyroid Function Parameters. Kathmandu Univ Med J (KUMJ). 2018;16(62):129-137.

5. Haskard-Zolnierek K, Wilson C, Pruin J, Deason R, Howard K. The Relationship Between Brain Fog and Medication Adherence for Individuals With Hypothyroidism. Clin Nurs Res. 2022;31(3):445-452. doi:10.1177/10547738211038127

6. McNally LJ, Ofiaeli CI, Oyibo SO. Treatment-refractory hypothyroidism. BMJ. 2019;364:l579. Published 2019 Feb 25. doi:10.1136/bmj.l579

7. Nakano Y, Hashimoto K, Ohkiba N, et al. A Case of Refractory Hypothyroidism due to Poor Compliance Treated with the Weekly Intravenous and Oral Levothyroxine Administration. Case Rep Endocrinol. 2019;2019:5986014. Published 2019 Feb 5. doi:10.1155/2019/5986014

8. Kiran Z, Shaikh KS, Fatima N, Tariq N, Baloch AA. Levothyroxine absorption test followed by directly observed treatment on an outpatient basis to address long-term high TSH levels in a hypothyroid patient: a case report. J Med Case Rep. 2023;17(1):24. Published 2023 Jan 25. doi:10.1186/s13256-023-03760-0

9. Chiu HH, Larrazabal R Jr, Uy AB, Jimeno C. Weekly Versus Daily Levothyroxine Tablet Replacement in Adults with Hypothyroidism: A Meta-Analysis. J ASEAN Fed Endocr Soc. 2021;36(2):156-160. doi:10.15605/jafes.036.02.07

10. Cappelli C, Castello R, Marini F, et al. Adherence to Levothyroxine Treatment Among Patients With Hypothyroidism: A Northeastern Italian Survey. Front Endocrinol (Lausanne). 2018;9:699. Published 2018 Nov 23. doi:10.3389/fendo.2018.00699

11. Bocale R, Desideri G, Barini A, et al. Long-Term Adherence to Levothyroxine Replacement Therapy in Thyroidectomized Patients. J Clin Med. 2022;11(15):4296. Published 2022 Jul 24. doi:10.3390/jcm11154296

12. El Helou S, Hallit S, Awada S, et al. Adherence to levothyroxine among patients with hypothyroidism in Lebanon. East Mediterr Health J. 2019;25(3):149-159. Published 2019 Apr 25. doi:10.26719/emhj.18.022

References

1. Chaker L, Bianco AC, Jonklaas J, Peeters RP. Hypothyroidism. Lancet. 2017;390(10101):1550-1562. doi:10.1016/S0140-6736(17)30703-1

2. Centanni M, Benvenga S, Sachmechi I. Diagnosis and management of treatment-refractory hypothyroidism: an expert consensus report. J Endocrinol Invest. 2017;40(12):1289-1301. doi:10.1007/s40618-017-0706-y

3. Hepp Z, Lage MJ, Espaillat R, Gossain VV. The association between adherence to levothyroxine and economic and clinical outcomes in patients with hypothyroidism in the US. J Med Econ. 2018;21(9):912-919. doi:10.1080/13696998.2018.1484749

4. Shakya Shrestha S, Risal K, Shrestha R, Bhatta RD. Medication Adherence to Levothyroxine Therapy among Hypothyroid Patients and their Clinical Outcomes with Special Reference to Thyroid Function Parameters. Kathmandu Univ Med J (KUMJ). 2018;16(62):129-137.

5. Haskard-Zolnierek K, Wilson C, Pruin J, Deason R, Howard K. The Relationship Between Brain Fog and Medication Adherence for Individuals With Hypothyroidism. Clin Nurs Res. 2022;31(3):445-452. doi:10.1177/10547738211038127

6. McNally LJ, Ofiaeli CI, Oyibo SO. Treatment-refractory hypothyroidism. BMJ. 2019;364:l579. Published 2019 Feb 25. doi:10.1136/bmj.l579

7. Nakano Y, Hashimoto K, Ohkiba N, et al. A Case of Refractory Hypothyroidism due to Poor Compliance Treated with the Weekly Intravenous and Oral Levothyroxine Administration. Case Rep Endocrinol. 2019;2019:5986014. Published 2019 Feb 5. doi:10.1155/2019/5986014

8. Kiran Z, Shaikh KS, Fatima N, Tariq N, Baloch AA. Levothyroxine absorption test followed by directly observed treatment on an outpatient basis to address long-term high TSH levels in a hypothyroid patient: a case report. J Med Case Rep. 2023;17(1):24. Published 2023 Jan 25. doi:10.1186/s13256-023-03760-0

9. Chiu HH, Larrazabal R Jr, Uy AB, Jimeno C. Weekly Versus Daily Levothyroxine Tablet Replacement in Adults with Hypothyroidism: A Meta-Analysis. J ASEAN Fed Endocr Soc. 2021;36(2):156-160. doi:10.15605/jafes.036.02.07

10. Cappelli C, Castello R, Marini F, et al. Adherence to Levothyroxine Treatment Among Patients With Hypothyroidism: A Northeastern Italian Survey. Front Endocrinol (Lausanne). 2018;9:699. Published 2018 Nov 23. doi:10.3389/fendo.2018.00699

11. Bocale R, Desideri G, Barini A, et al. Long-Term Adherence to Levothyroxine Replacement Therapy in Thyroidectomized Patients. J Clin Med. 2022;11(15):4296. Published 2022 Jul 24. doi:10.3390/jcm11154296

12. El Helou S, Hallit S, Awada S, et al. Adherence to levothyroxine among patients with hypothyroidism in Lebanon. East Mediterr Health J. 2019;25(3):149-159. Published 2019 Apr 25. doi:10.26719/emhj.18.022

Issue
Federal Practitioner - 41(3)a
Issue
Federal Practitioner - 41(3)a
Page Number
84
Page Number
84
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>0324 FED Hypothyroidism</fileName> <TBEID>0C02F244.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F244</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240302T213953</firstPublished> <LastPublished>20240302T213953</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240302T213953</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Sabrina Kaul;a,b Ankur Gupta, MDa,b</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>Nonadherence to medications is an issue across health care. In endocrinology, hypothyroidism, a deficiency of thyroid hormones, is most often treated with levot</metaDescription> <articlePDF/> <teaserImage/> <title>Medication-Nonadherent Hypothyroidism Requiring Frequent Primary Care Visits to Achieve Euthyroidism</title> <deck/> <eyebrow>Case in Point</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>March</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2953</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>March 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case in Point | 2953<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">45</term> </sections> <topics> <term canonical="true">27442</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Medication-Nonadherent Hypothyroidism Requiring Frequent Primary Care Visits to Achieve Euthyroidism</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Nonadherence to medications is a common clinical issue. We describe a case in which weekly visits to assess medication adherence achieved euthyroidism in a patient with persistent primary hypothyroidism and suspected nonadherence to levothyroxine. The patient, however, did not report nonadherence.<br/><br/><b>Case Presentation:</b> A male aged 67 years with multinodular goiter underwent total thyroidectomy for abnormal thyroid nodule biopsy. Surgical pathology revealed papillary thyroid cancer with lymph node metastasis for which he received radioactive iodine treatment. His plasma thyrotropin (TSH) was noted to be 0.28 uIU/mL (reference range 0.35-4.00 uIU/mL) 7 months postsurgery while taking 224 mcg levothyroxine tablets daily. His plasma TSH remained elevated for about 5 years despite titrations of the levothyroxine dosage, counseling, and multiple follow-up visits. A home care nurse was involved in monitoring the patient taking levothyroxine daily and correctly but was unsuccessful. The patient and his son reported taking levothyroxine daily and correctly. The patient was asked to visit the primary care clinic every week for 6 weeks with all his medications. Repeat plasma TSH normalized to 1.01 uIU/mL. The suspected etiology of previously high plasma TSH was nonadherence to levothyroxine, which was discussed in detail with the patient. The patient verbalized understanding, was willing to follow recommendations and ended the weekly clinic visits. Repeat plasma TSH was again high and the patient claimed adherence, but weekly visits to primary care clinic were resumed, and life-threatening consequences of hypothyroidism were discussed with the patient and his son. After 9 weeks of visits, he was noted to have low plasma TSH (0.23 uIU/mL).<br/><br/><b>Conclusions:</b> Weekly visits seem impractical but may help in cases of persistent hypothyroidism in which the patient admits to being or is suspected to be nonadherent to levothyroxine. Knowing their medication use will be checked at weekly clinic visits may motivate the patient to be adherent.</p> <p><span class="Drop">N</span>onadherence to medications is an issue across health care. In endocrinology, hypothyroidism, a deficiency of thyroid hormones, is most often treated with levothyroxine and if left untreated can lead to myxedema coma, which can lead to death due to multiorgan dysfunction.<sup>1</sup> Therefore, adherence to levothyroxine is very important in preventing fatal complications. </p> <p>We present the case of a patient with persistent primary hypothyroidism who was suspected to be nonadherent to levothyroxine, although the patient consistently claimed adherence. The patient’s plasma thyrotropin (TSH) level improved to reference range after 6 weeks of weekly primary care clinic visits. After stopping the visits, his plasma TSH level increased again, so 9 more weeks of visits resumed, which again helped bring down his plasma TSH levels.</p> <h2>Case Presentation</h2> <p>A male patient aged 67 years presented to the Dayton Veterans Affairs Medical Center (VAMC) endocrinology clinic for evaluation of thyroid nodules. The patient reported no history of neck irradiation and a physical examination was unremarkable. At that time, laboratory results showed a slightly elevated plasma TSH level of 4.35 uIU/mL (reference range, 0.35-4.00 uIU/mL) and normal free thyroxine (T4) of 1.00 ng/dL (reference range, 0.74-1.46 ng/dL). Later that year, the patient underwent a total thyroidectomy at the Cincinnati VAMC for Hurthle cell variant papillary thyroid carcinoma that was noted on biopsy at the Dayton VAMC. After surgical pathology results were available, the patient started levothyroxine 200 mcg daily, although 224 mcg would have been more appropriate based on his 142 kg weight. Due to a history of arrhythmia, the goal plasma TSH level was 0.10 to 0.50 uIU/mL. The patient subsequently underwent radioactive iodine ablation. After levothyroxine dose adjustments, the patient’s plasma TSH level was noted to be within his target range at 0.28 uIU/mL 3 months postablation. </p> <p>Over the next 5 years the patient had regular laboratory tests during which his plasma TSH level rose and were typically high despite adjusting levothyroxine doses between 200 mcg and 325 mcg. The patient received counseling on taking the medication in the morning on an empty stomach and waiting at least 1 hour before consuming anything, and he went to many follow-up visits at the Dayton VAMC endocrinology clinic<b>.</b> He reported no vomiting or diarrhea but endorsed weight gain once. The patient also had high free T4 at times and did not take extra levothyroxine before undergoing laboratory tests. <br/><br/>Nonadherence to levothyroxine was suspected, but the patient insisted he was adherent. He received the medication in the mail regularly, generally had 90-day refills unless a dose change was made, used a pill box, and had social support from his son, but he did not use a phone alarm to remind him to take it. A home care nurse made weekly visits to make sure the remaining levothyroxine pill counts were correct; however, the patient continued to have difficulty maintaining daily adherence at home as indicated by the nurse’s pill counts not aligning with the number of pills which should have been left if the patient was talking the pills daily. <br/><br/>The patient was asked to visit a local community-based outpatient clinic (CBOC) weekly (to avoid patient travel time to Dayton VAMC &gt; 1 hour) to check pill counts and assess adherence. The patient went to the CBOC clinic for these visits, during which pill counts indicated much better but not 100% adherence. After 6 weeks of clinic visits, his plasma TSH decreased to 1.01 uIU/mL, which was within the reference range, and the patient stopped coming to the weekly clinic visits (Table). Four months later, the patient's plasma TSH levels increased to 80.72 uIU/mL. Nonadherence to levothyroxine was suspected again. He was asked to resume weekly clinic visits, and the life-threatening effects of hypothyroidism and not taking levothyroxine were discussed with the patient and his son. The patient made CBOC clinic visits for 9 weeks, after which his plasma TSH level was low at 0.23 uIU/mL.</p> <h2>Discussion</h2> <p>There are multiple important causes to consider in patients with persistent hypothyroidism. One is medication nonadherence, which was most likely seen in the patient in this case. Missing even 1 day of levothyroxine can affect TSH and thyroid hormone levels for several days due to the long half-life of the medication.<sup>2</sup> Hepp and colleagues found that patients with hypothyroidism were significantly more likely to be nonadherent to levothyroxine if they had comorbid conditions such as type 2 diabetes or were obese.<sup>3</sup> Another study of levothyroxine adherence found that the most common reason for missing doses was forgetfulness.<sup>4</sup> However, memory and cognition impairments can also be symptoms of hypothyroidism itself; Haskard-Zolnierek and colleagues found a significant association between nonadherence to levothyroxine and self-reported brain fog in patients with hypothyroidism.<sup>5</sup></p> <p>Another cause of persistent hypothyroidism is malabsorption. Absorption of levothyroxine can be affected by intestinal malabsorption due to inflammatory bowel disease, lactose intolerance, or gastrointestinal infection, as well as several foods, drinks (eg, coffee), medications, vitamins, and supplements (eg, proton-pump inhibitors and calcium).<sup>2,6</sup> Levothyroxine is absorbed mainly at the jejunum and upper ileum, so any pathologies or ingested items that would directly or indirectly affect absorption at those sites can affect levothyroxine absorption.<sup>2</sup> <br/><br/>A liquid levothyroxine formulation can help with malabsorption.<sup>2</sup> Alternatively, weight gain may lead to a need for increasing the dosage of levothyroxine.<sup>2,6</sup> Other factors that can affect TSH levels include Addison disease, dysregulation of the hypothalamic-pituitary-thyroid axis, and TSH heterophile antibodies.<sup>2<br/><br/></sup>Research describes methods that have effectively treated hypothyroidism in patients struggling with levothyroxine adherence. Two case reports describe weekly visits for levothyroxine administration successfully treating uncontrolled hypothyroidism.<sup>7,8 </sup>A meta-analysis found that while weekly levothyroxine tablets led to a higher mean TSH level than daily use, weekly use still led to reference-range TSH levels, suggesting that weekly levothyroxine may be a helpful alternative for nonadherent patients.<sup>9</sup> Alternatively, patients taking levothyroxine tablets have been shown to forget to take their medication more frequently compared to those taking the liquid formulation.<sup>10,11</sup> Additionally, a study by El Helou and colleagues found that adherence to levothyroxine was significantly improved when patients had endocrinology visits once a month and when the endocrinologist provided information about hypothyroidism.<sup>12<br/><br/></sup>Another method that may improve adherence to levothyroxine is telehealth visits. This would be especially helpful for patients who live far from the clinic or do not have the time, transportation, or financial means to visit the clinic for weekly visits to assess medication adherence. Additionally, patients may be afraid of admitting to a health care professional that they are nonadherent. Clinicians must be tactful when asking about adherence to make the patient feel comfortable with admitting to nonadherence if their cognition is not impaired. Then, a patient-led conversation can occur regarding realistic ways the patient feels they can work toward adherence.<br/><br/>To our knowledge, the patient in this case report had no symptoms of intestinal malabsorption, and weight gain was not thought to be the issue, as levothyroxine dosage was adjusted multiple times. His plasma TSH levels returned to reference range after weekly pill count visits for 6 weeks and after weekly pill count visits for 9 weeks. Therefore, nonadherence to levothyroxine was suspected to be the cause of frequently elevated plasma TSH levels despite the patient’s insistence on adherence. While the patient did not report memory issues, cognitive impairments due to hypothyroidism may have been contributing to his probable nonadherence. Additionally, he had comorbidities, such as type 2 diabetes mellitus and obesity, which may have made adherence more difficult. <br/><br/>Levothyroxine was also only prescribed in daily tablet form, so the frequency and formulation may have also contributed to nonadherence. While the home nurse was originally sent to assess the patient’s adherence, the care team could have had the nurse start giving the patient weekly levothyroxine once nonadherence was determined to be a likely issue. The patient’s adherence only improved when he went to the clinic for pill counts but not when the home nurse came to his house weekly; this could be because the patient knew he had to invest the time to physically go to clinic visits for pill checks, motivating him to increase adherence.</p> <h2>Conclusions</h2> <p>This case reports a patient with frequently high plasma TSH levels achieving normalization of plasma TSH levels after weekly medication adherence checks at a primary care clinic. Weekly visits to a clinic seem impractical compared to weekly dosing with a visiting nurse; however, after review of the literature, this may be an approach to consider in the future. This strategy may especially help in cases of persistent abnormal plasma TSH levels in which no etiology can be found other than suspected medication nonadherence. Knowing their medication use will be checked at weekly clinic visits may motivate patients to be adherent.</p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Dayton Veterans Affairs Medical Center, Ohio<br/><br/><sup>b</sup>Wright State University Boonshoft School of Medicine, Dayton, Ohio</em> </p> <p class="isub">Author disclosures</p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <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 drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.</em> </p> <p class="isub">Ethics and consent </p> <p> <em>Written informed consent was obtained from the patient presented in this case report. Patient identifiers have been removed to protect the privacy of the patient.</em> </p> <p class="isub">References</p> <p class="reference"> 1. Chaker L, Bianco AC, Jonklaas J, Peeters RP. Hypothyroidism. <i>Lancet</i>. 2017;390(10101):1550-1562. doi:10.1016/S0140-6736(17)30703-1<br/><br/> 2. Centanni M, Benvenga S, Sachmechi I. Diagnosis and management of treatment-refractory hypothyroidism: an expert consensus report. <i>J Endocrinol Invest</i>. 2017;40(12):1289-1301. doi:10.1007/s40618-017-0706-y <br/><br/> 3. Hepp Z, Lage MJ, Espaillat R, Gossain VV. The association between adherence to levothyroxine and economic and clinical outcomes in patients with hypothyroidism in the US. <i>J Med Econ</i>. 2018;21(9):912-919. doi:10.1080/13696998.2018.1484749<br/><br/> 4. Shakya Shrestha S, Risal K, Shrestha R, Bhatta RD. Medication Adherence to Levothyroxine Therapy among Hypothyroid Patients and their Clinical Outcomes with Special Reference to Thyroid Function Parameters. <i>Kathmandu Univ Med J (KUMJ)</i>. 2018;16(62):129-137.<br/><br/> 5. Haskard-Zolnierek K, Wilson C, Pruin J, Deason R, Howard K. The Relationship Between Brain Fog and Medication Adherence for Individuals With Hypothyroidism. <i>Clin Nurs Res</i>. 2022;31(3):445-452. doi:10.1177/10547738211038127<br/><br/> 6. McNally LJ, Ofiaeli CI, Oyibo SO. Treatment-refractory hypothyroidism. <i>BMJ</i>. 2019;364:l579. Published 2019 Feb 25. doi:10.1136/bmj.l579<br/><br/> 7. Nakano Y, Hashimoto K, Ohkiba N, et al. A Case of Refractory Hypothyroidism due to Poor Compliance Treated with the Weekly Intravenous and Oral Levothyroxine Administration. <i>Case Rep Endocrinol</i>. 2019;2019:5986014. Published 2019 Feb 5. doi:10.1155/2019/5986014<br/><br/> 8. Kiran Z, Shaikh KS, Fatima N, Tariq N, Baloch AA. Levothyroxine absorption test followed by directly observed treatment on an outpatient basis to address long-term high TSH levels in a hypothyroid patient: a case report. <i>J Med Case Rep</i>. 2023;17(1):24. Published 2023 Jan 25. doi:10.1186/s13256-023-03760-0<br/><br/> 9. Chiu HH, Larrazabal R Jr, Uy AB, Jimeno C. Weekly Versus Daily Levothyroxine Tablet Replacement in Adults with Hypothyroidism: A Meta-Analysis. <i>J ASEAN Fed Endocr Soc</i>. 2021;36(2):156-160. doi:10.15605/jafes.036.02.07<br/><br/>10. Cappelli C, Castello R, Marini F, et al. Adherence to Levothyroxine Treatment Among Patients With Hypothyroidism: A Northeastern Italian Survey. <i>Front Endocrinol (Lausanne)</i>. 2018;9:699. Published 2018 Nov 23. doi:10.3389/fendo.2018.00699<br/><br/>11. Bocale R, Desideri G, Barini A, et al. Long-Term Adherence to Levothyroxine Replacement Therapy in Thyroidectomized Patients. <i>J Clin Med</i>. 2022;11(15):4296. Published 2022 Jul 24. doi:10.3390/jcm11154296<br/><br/>12. El Helou S, Hallit S, Awada S, et al. Adherence to levothyroxine among patients with hypothyroidism in Lebanon. <i>East Mediterr Health J</i>. 2019;25(3):149-159. Published 2019 Apr 25. doi:10.26719/emhj.18.022</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

Dermatologic Reactions Following COVID-19 Vaccination: A Case Series

Article Type
Changed
Thu, 02/22/2024 - 10:06
Display Headline
Dermatologic Reactions Following COVID-19 Vaccination: A Case Series

Cutaneous reactions associated with the Pfizer-BioNTech COVID-19 vaccine have been reported worldwide since December 2020. Local injection site reactions (<1%) such as erythema, swelling, delayed local reactions (1%–10%), morbilliform rash, urticarial reactions, pityriasis rosea, Rowell syndrome, and lichen planus have been reported following the Pfizer-BioNTech COVID-19 vaccine.1 Cutaneous reactions reported in association with the Sinovac-Coronavac COVID-19 vaccine include swelling, redness, itching, discoloration, induration (1%–10%), urticaria, petechial rash, and exacerbation of psoriasis at the local injection site (<1%).2

We describe 7 patients from Turkey who presented with various dermatologic problems 5 to 28 days after COVID-19 vaccination, highlighting the possibility of early and late cutaneous reactions related to the vaccine (Table).

CT11302016_e_Table.jpg

Case Reports

Patient 1—A 44-year-old woman was admitted to the dermatology clinic with painful lesions on the trunk of 3 days’ duration. Dermatologic examination revealed grouped erythematous vesicles showing dermatomal spread in the right thoracolumbar (dermatome T10) region. The patient reported that she had received 2 doses of the Sinovac-Coronavac vaccine (doses 1 and 2) and 2 doses of the BioNTech COVID-19 vaccine (doses 3 and 4); the rash had developed 28 days after she received the 4th dose. Her medical history was unremarkable. The lesions regressed after 1 week of treatment with oral valacyclovir 1000 mg 3 times daily, but she developed postherpetic neuralgia 1 week after starting treatment, which resolved after 8 weeks.

Patient 2—A 68-year-old woman presented to the dermatology clinic for evaluation of painful sores on the upper lip of 1 day’s duration. She had a history of rheumatoid arthritis, hypertension, and atopy and was currently taking prednisone and etanercept. Dermatologic examination revealed grouped vesicles on an erythematous base on the upper lip. A diagnosis of herpes labialis was made. The patient reported that she had received a third dose of the Sinovac-Coronavac vaccine 10 days prior to the appearance of the lesions. Her symptoms resolved completely within 2 weeks of treatment with topical acyclovir.

Patient 3—A 64-year-old woman was admitted to the hospital with pain, redness, and watery sores on and around the left eyelid of 2 days’ duration. Dermatologic evaluation revealed the erythematous surface of the left eyelid and periorbital area showed partial crusts, clustered vesicles, erythema, and edema. Additionally, the conjunctiva was purulent and erythematous. The patient’s medical history was notable for allergic asthma, hypertension, anxiety, and depression. For this reason, the patient was prescribed an angiotensin receptor blocker and a selective serotonin reuptake inhibitor. She noted that a similar rash had developed around the left eye 6 years prior that was diagnosed as herpes zoster (HZ). She also reported that she had received 2 doses of the Sinovac-Coronavac COVID-19 vaccine followed by 1 dose of the BioNTech COVID-19 vaccine, which she had received 2 weeks before the rash developed. The patient was treated at the eye clinic and was found to have ocular involvement. Ophthalmology was consulted and a diagnosis of herpes zoster ophthalmicus (HZO) was made. Systemic valacyclovir treatment was initiated, resulting in clinical improvement within 3 weeks.

Patient 4—A 75-year-old man was admitted to the hospital with chest and back pain and widespread muscle pain of several days’ duration. His medical history was remarkable for diabetes mellitus, hypertension, depression, and coronary artery bypass surgery. A medication history revealed treatment with a β-blocker, acetylsalicylic acid, a calcium channel blocker, a dipeptidyl peptidase 4 inhibitor, and a selective serotonin reuptake inhibitor. Dermatologic examination revealed grouped vesicles on an erythematous background in dermatome T5 on the right chest and back. A diagnosis of HZ was made. The patient reported that he had received 2 doses of the Sinovac-Coronavac vaccine followed by 1 dose of the Pfizer-BioNTech vaccine 2 weeks prior to the current presentation. He was treated with valacyclovir for 1 week, and his symptoms resolved entirely within 3 weeks.

Patient 5—A 50-year-old woman presented to the hospital for evaluation of painful sores on the back, chest, groin, and abdomen of 10 days’ duration. The lesions initially had developed 7 days after receiving the BioNTech COVID-19 vaccine; she previously had received 2 doses of the Sinovac-Coronavac vaccine. The patient had a history of untreated psoriasis. Dermatologic examination revealed grouped vesicles on an erythematous background in the T2–L2 dermatomes on the left side of the trunk. A diagnosis of HZ was made. The lesions resolved after 1 week of treatment with systemic valacyclovir; however, she subsequently developed postherpetic neuralgia, hypoesthesia, and postinflammatory hyperpigmentation in the affected regions.

 

 

Patient 6—A 37-year-old woman presented to the hospital with redness, swelling, and itching all over the body of 3 days’ duration. The patient noted that the rash would subside and reappear throughout the day. Her medical history was unremarkable, except for COVID-19 infection 6 months prior. She had received a second dose of the BioNTech vaccine 20 days prior to development of symptoms. Dermatologic examination revealed widespread erythematous urticarial plaques. A diagnosis of acute urticaria was made. The patient recovered completely after 1 week of treatment with a systemic steroid and 3 weeks of antihistamine treatment.

Patient 7—A 63-year-old woman presented to the hospital with widespread itching and rash that appeared 5 days after the first dose of the BioNTech COVID-19 vaccine. The patient reported that the rash resolved spontaneously within a few hours but then reappeared. Her medical history revealed that she was taking tamoxifen for breast cancer and that she previously had received 2 doses of the Sinovac-Coronavac vaccine. Dermatologic examination revealed erythematous urticarial plaques on the trunk and arms. A diagnosis of urticaria was made, and her symptoms resolved after 6 weeks of antihistamine treatment.

Comment

Skin lesions associated with COVID-19 infection have been reported worldwide3,4 as well as dermatologic reactions following COVID-19 vaccination. In one case from Turkey, HZ infection was reported in a 68-year-old man 5 days after he received a second dose of the COVID-19 vaccine.5 In another case, HZ infection developed in a 78-year-old man 5 days after COVID-19 vaccination.6 Numerous cases of HZ infection developing within 1 to 26 days of COVID-19 vaccination have been reported worldwide.7-9

In a study conducted in the United States, 40 skin reactions associated with the COVID-19 vaccine were investigated; of these cases, 87.5% (35/40) were reported as varicella-zoster virus, and 12.5% (5/40) were reported as herpes simplex reactivation; 54% (19/35) and 80% (4/5) of these cases, respectively, were associated with the Pfizer-BioNTech vaccine.10 The average age of patients who developed a skin reaction was 46 years, and 70% (28/40) were women. The time to onset of the reaction was 2 to 13 days after vaccination, and symptoms were reported to improve within 7 days on average.10

Another study from Spain examined 405 vaccine-related skin reactions, 40.2% of which were related to the Pfizer-BioNTech vaccine. Among them, 80.2% occurred in women; 13.8% of cases were diagnosed as varicella-zoster virus or HZ virus reactivation, and 14.6% were urticaria. Eighty reactions (21%) were classified as severe/very severe and 81% required treatment.11 One study reported 414 skin reactions from the COVID-19 vaccine from December 2020 to February 2021; of these cases, 83% occurred after the Moderna vaccine, which is not available in Turkey, and 17% occurred after the Pfizer-BioNTech vaccine.12A systematic review of 91 patients who developed HZ infection after COVID-19 vaccination reported that 10% (9/91) of cases were receiving immunosuppressive therapy and 13% (12/91) had an autoimmune disease.7 In our case series, it is known that at least 2 of the patients (patients 2 and 5), including 1 patient with rheumatoid arthritis (patient 2) who was on immunosuppressive treatment, had autoimmune disorders. However, reports in the literature indicate that most patients with autoimmune inflammatory rheumatic diseases remain stable after vaccination.13

Herpes zoster ophthalmicus is a rare form of HZ caused by involvement of the ophthalmic branch of the trigeminal nerve that manifests as vesicular lesions and retinitis, uveitis, keratitis, conjunctivitis, and pain on an erythematous background. Two cases of women who developed HZO infection after Pfizer-BioNTech vaccination were reported in the literature.14 Although patient 3 in our case series had a history of HZO 6 years prior, the possibility of the Pfizer-BioNTech vaccine triggering HZO should be taken into consideration.

Although cutaneous reactions after the Sinovac-Coronavac vaccine were observed in only 1 of 7 patients in our case series, skin reactions after Sinovac-Coronavac (an inactivated viral vaccine) have been reported in the literature. In one study, after a total of 35,229 injections, the incidence of cutaneous adverse events due to Sinovac-Coronavac was reported to be 0.94% and 0.70% after the first and second doses, respectively.15 Therefore, further study results are needed to directly attribute the reactions to COVID-19 vaccination.

Conclusion

Our case series highlights that clinicians should be vigilant in diagnosing cutaneous reactions following COVID-19 vaccination early to prevent potential complications. Early recognition of reactions is crucial, and the prognosis can be improved with appropriate treatment. Despite the potential dermatologic adverse effects of the COVID-19 vaccine, the most effective way to protect against serious COVID-19 infection is to continue to be vaccinated.

References
  1. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N Engl J Med. 2020;383:2603-2615.
  2. Zhang Y, Zeng G, Pan H, et al. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18–59 years: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect Dis. 2021;21:181-192.
  3. Tan SW, Tam YC, Oh CC. Skin manifestations of COVID-19: a worldwide review. JAAD Int. 2021;2:119-133.
  4. Singh H, Kaur H, Singh K, et al. Cutaneous manifestations of COVID-19: a systematic review. advances in wound care. 2021;10:51-80.
  5. Aksu SB, Öztürk GZ. A rare case of shingles after COVID-19 vaccine: is it a possible adverse effect? clinical and experimental vaccine research. 2021;10:198-201.
  6. Bostan E, Yalici-Armagan B. Herpes zoster following inactivated COVID-19 vaccine: a coexistence or coincidence? J Cosmet Dermatol. 2021;20:1566-1567.
  7. Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. Vaccines (Basel). 2021;9:1013. doi:10.3390/vaccines9091013
  8. Rodríguez-Jiménez P, Chicharro P, Cabrera LM, et al. Varicella-zoster virus reactivation after SARS-CoV-2 BNT162b2 mRNA vaccination: report of 5 cases. JAAD Case Rep. 2021;12:58-59. doi:10.1016/j.jdcr.2021.04.014
  9. Lee C, Cotter D, Basa J, et al. 20 Post-COVID-19 vaccine-related shingles cases seen at the Las Vegas Dermatology clinic and sent to us via social media. J Cosmet Dermatol. 2021;20:1960-1964.
  10. Fathy RA, McMahon DE, Lee C, et al. Varicella-zoster and herpes simplex virus reactivation post-COVID-19 vaccination: a review of 40 cases in an International Dermatology Registry. J Eur Acad Dermatol Venerol. 2022;36:E6-E9.
  11. Català A, Muñoz-Santos C, Galván-Casas C, et al. Cutaneous reactions after SARS-CoV-2 vaccination: a cross-sectional Spanish nationwide study of 405 cases. Br J Dermatol. 2022;186:142-152.
  12. McMahon DE, Amerson E, Rosenbach M, et al. Cutaneous reactions reported after Moderna and Pfizer COVID-19 vaccination: a registry-based study of 414 cases. J Am Acad Dermatol. 2021;85:46-55.
  13. Furer V, Eviatar T, Zisman D, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. Ann Rheum Dis. 2021;80:1330-1338.
  14. Bernardini N, Skroza N, Mambrin A, et al. Herpes zoster ophthalmicus in two women after Pfizer-BioNTech (BNT162b2) vaccine. J Med Virol. 2022;94:817-818.
  15. Rerknimitr P, Puaratanaarunkon T, Wongtada C, et al. Cutaneous adverse reactions from 35,229 doses of Sinovac and AstraZeneca COVID-19 vaccination: a prospective cohort study in healthcare workers. J Eur Acad Dermatol Venereol. 2022;36:E158-E161.
Article PDF
Author and Disclosure Information

From Giresun University, Turkey. Dr. Alkanat is from the Faculty of Health Sciences, and Dr. Kulaklı is from the Faculty of Medicine, Dermatology Clinic.

The authors report no conflict of interest.

Correspondence: Hafize Özdemir Alkanat, RN, PhD, Giresun University, Faculty of Health Sciences, 28340 Piraziz/Giresun, Turkey (hafize.ozdemir@giresun.edu.tr).

Issue
Cutis - 113(2)
Publications
Topics
Page Number
E16-E19
Sections
Author and Disclosure Information

From Giresun University, Turkey. Dr. Alkanat is from the Faculty of Health Sciences, and Dr. Kulaklı is from the Faculty of Medicine, Dermatology Clinic.

The authors report no conflict of interest.

Correspondence: Hafize Özdemir Alkanat, RN, PhD, Giresun University, Faculty of Health Sciences, 28340 Piraziz/Giresun, Turkey (hafize.ozdemir@giresun.edu.tr).

Author and Disclosure Information

From Giresun University, Turkey. Dr. Alkanat is from the Faculty of Health Sciences, and Dr. Kulaklı is from the Faculty of Medicine, Dermatology Clinic.

The authors report no conflict of interest.

Correspondence: Hafize Özdemir Alkanat, RN, PhD, Giresun University, Faculty of Health Sciences, 28340 Piraziz/Giresun, Turkey (hafize.ozdemir@giresun.edu.tr).

Article PDF
Article PDF

Cutaneous reactions associated with the Pfizer-BioNTech COVID-19 vaccine have been reported worldwide since December 2020. Local injection site reactions (<1%) such as erythema, swelling, delayed local reactions (1%–10%), morbilliform rash, urticarial reactions, pityriasis rosea, Rowell syndrome, and lichen planus have been reported following the Pfizer-BioNTech COVID-19 vaccine.1 Cutaneous reactions reported in association with the Sinovac-Coronavac COVID-19 vaccine include swelling, redness, itching, discoloration, induration (1%–10%), urticaria, petechial rash, and exacerbation of psoriasis at the local injection site (<1%).2

We describe 7 patients from Turkey who presented with various dermatologic problems 5 to 28 days after COVID-19 vaccination, highlighting the possibility of early and late cutaneous reactions related to the vaccine (Table).

CT11302016_e_Table.jpg

Case Reports

Patient 1—A 44-year-old woman was admitted to the dermatology clinic with painful lesions on the trunk of 3 days’ duration. Dermatologic examination revealed grouped erythematous vesicles showing dermatomal spread in the right thoracolumbar (dermatome T10) region. The patient reported that she had received 2 doses of the Sinovac-Coronavac vaccine (doses 1 and 2) and 2 doses of the BioNTech COVID-19 vaccine (doses 3 and 4); the rash had developed 28 days after she received the 4th dose. Her medical history was unremarkable. The lesions regressed after 1 week of treatment with oral valacyclovir 1000 mg 3 times daily, but she developed postherpetic neuralgia 1 week after starting treatment, which resolved after 8 weeks.

Patient 2—A 68-year-old woman presented to the dermatology clinic for evaluation of painful sores on the upper lip of 1 day’s duration. She had a history of rheumatoid arthritis, hypertension, and atopy and was currently taking prednisone and etanercept. Dermatologic examination revealed grouped vesicles on an erythematous base on the upper lip. A diagnosis of herpes labialis was made. The patient reported that she had received a third dose of the Sinovac-Coronavac vaccine 10 days prior to the appearance of the lesions. Her symptoms resolved completely within 2 weeks of treatment with topical acyclovir.

Patient 3—A 64-year-old woman was admitted to the hospital with pain, redness, and watery sores on and around the left eyelid of 2 days’ duration. Dermatologic evaluation revealed the erythematous surface of the left eyelid and periorbital area showed partial crusts, clustered vesicles, erythema, and edema. Additionally, the conjunctiva was purulent and erythematous. The patient’s medical history was notable for allergic asthma, hypertension, anxiety, and depression. For this reason, the patient was prescribed an angiotensin receptor blocker and a selective serotonin reuptake inhibitor. She noted that a similar rash had developed around the left eye 6 years prior that was diagnosed as herpes zoster (HZ). She also reported that she had received 2 doses of the Sinovac-Coronavac COVID-19 vaccine followed by 1 dose of the BioNTech COVID-19 vaccine, which she had received 2 weeks before the rash developed. The patient was treated at the eye clinic and was found to have ocular involvement. Ophthalmology was consulted and a diagnosis of herpes zoster ophthalmicus (HZO) was made. Systemic valacyclovir treatment was initiated, resulting in clinical improvement within 3 weeks.

Patient 4—A 75-year-old man was admitted to the hospital with chest and back pain and widespread muscle pain of several days’ duration. His medical history was remarkable for diabetes mellitus, hypertension, depression, and coronary artery bypass surgery. A medication history revealed treatment with a β-blocker, acetylsalicylic acid, a calcium channel blocker, a dipeptidyl peptidase 4 inhibitor, and a selective serotonin reuptake inhibitor. Dermatologic examination revealed grouped vesicles on an erythematous background in dermatome T5 on the right chest and back. A diagnosis of HZ was made. The patient reported that he had received 2 doses of the Sinovac-Coronavac vaccine followed by 1 dose of the Pfizer-BioNTech vaccine 2 weeks prior to the current presentation. He was treated with valacyclovir for 1 week, and his symptoms resolved entirely within 3 weeks.

Patient 5—A 50-year-old woman presented to the hospital for evaluation of painful sores on the back, chest, groin, and abdomen of 10 days’ duration. The lesions initially had developed 7 days after receiving the BioNTech COVID-19 vaccine; she previously had received 2 doses of the Sinovac-Coronavac vaccine. The patient had a history of untreated psoriasis. Dermatologic examination revealed grouped vesicles on an erythematous background in the T2–L2 dermatomes on the left side of the trunk. A diagnosis of HZ was made. The lesions resolved after 1 week of treatment with systemic valacyclovir; however, she subsequently developed postherpetic neuralgia, hypoesthesia, and postinflammatory hyperpigmentation in the affected regions.

 

 

Patient 6—A 37-year-old woman presented to the hospital with redness, swelling, and itching all over the body of 3 days’ duration. The patient noted that the rash would subside and reappear throughout the day. Her medical history was unremarkable, except for COVID-19 infection 6 months prior. She had received a second dose of the BioNTech vaccine 20 days prior to development of symptoms. Dermatologic examination revealed widespread erythematous urticarial plaques. A diagnosis of acute urticaria was made. The patient recovered completely after 1 week of treatment with a systemic steroid and 3 weeks of antihistamine treatment.

Patient 7—A 63-year-old woman presented to the hospital with widespread itching and rash that appeared 5 days after the first dose of the BioNTech COVID-19 vaccine. The patient reported that the rash resolved spontaneously within a few hours but then reappeared. Her medical history revealed that she was taking tamoxifen for breast cancer and that she previously had received 2 doses of the Sinovac-Coronavac vaccine. Dermatologic examination revealed erythematous urticarial plaques on the trunk and arms. A diagnosis of urticaria was made, and her symptoms resolved after 6 weeks of antihistamine treatment.

Comment

Skin lesions associated with COVID-19 infection have been reported worldwide3,4 as well as dermatologic reactions following COVID-19 vaccination. In one case from Turkey, HZ infection was reported in a 68-year-old man 5 days after he received a second dose of the COVID-19 vaccine.5 In another case, HZ infection developed in a 78-year-old man 5 days after COVID-19 vaccination.6 Numerous cases of HZ infection developing within 1 to 26 days of COVID-19 vaccination have been reported worldwide.7-9

In a study conducted in the United States, 40 skin reactions associated with the COVID-19 vaccine were investigated; of these cases, 87.5% (35/40) were reported as varicella-zoster virus, and 12.5% (5/40) were reported as herpes simplex reactivation; 54% (19/35) and 80% (4/5) of these cases, respectively, were associated with the Pfizer-BioNTech vaccine.10 The average age of patients who developed a skin reaction was 46 years, and 70% (28/40) were women. The time to onset of the reaction was 2 to 13 days after vaccination, and symptoms were reported to improve within 7 days on average.10

Another study from Spain examined 405 vaccine-related skin reactions, 40.2% of which were related to the Pfizer-BioNTech vaccine. Among them, 80.2% occurred in women; 13.8% of cases were diagnosed as varicella-zoster virus or HZ virus reactivation, and 14.6% were urticaria. Eighty reactions (21%) were classified as severe/very severe and 81% required treatment.11 One study reported 414 skin reactions from the COVID-19 vaccine from December 2020 to February 2021; of these cases, 83% occurred after the Moderna vaccine, which is not available in Turkey, and 17% occurred after the Pfizer-BioNTech vaccine.12A systematic review of 91 patients who developed HZ infection after COVID-19 vaccination reported that 10% (9/91) of cases were receiving immunosuppressive therapy and 13% (12/91) had an autoimmune disease.7 In our case series, it is known that at least 2 of the patients (patients 2 and 5), including 1 patient with rheumatoid arthritis (patient 2) who was on immunosuppressive treatment, had autoimmune disorders. However, reports in the literature indicate that most patients with autoimmune inflammatory rheumatic diseases remain stable after vaccination.13

Herpes zoster ophthalmicus is a rare form of HZ caused by involvement of the ophthalmic branch of the trigeminal nerve that manifests as vesicular lesions and retinitis, uveitis, keratitis, conjunctivitis, and pain on an erythematous background. Two cases of women who developed HZO infection after Pfizer-BioNTech vaccination were reported in the literature.14 Although patient 3 in our case series had a history of HZO 6 years prior, the possibility of the Pfizer-BioNTech vaccine triggering HZO should be taken into consideration.

Although cutaneous reactions after the Sinovac-Coronavac vaccine were observed in only 1 of 7 patients in our case series, skin reactions after Sinovac-Coronavac (an inactivated viral vaccine) have been reported in the literature. In one study, after a total of 35,229 injections, the incidence of cutaneous adverse events due to Sinovac-Coronavac was reported to be 0.94% and 0.70% after the first and second doses, respectively.15 Therefore, further study results are needed to directly attribute the reactions to COVID-19 vaccination.

Conclusion

Our case series highlights that clinicians should be vigilant in diagnosing cutaneous reactions following COVID-19 vaccination early to prevent potential complications. Early recognition of reactions is crucial, and the prognosis can be improved with appropriate treatment. Despite the potential dermatologic adverse effects of the COVID-19 vaccine, the most effective way to protect against serious COVID-19 infection is to continue to be vaccinated.

Cutaneous reactions associated with the Pfizer-BioNTech COVID-19 vaccine have been reported worldwide since December 2020. Local injection site reactions (<1%) such as erythema, swelling, delayed local reactions (1%–10%), morbilliform rash, urticarial reactions, pityriasis rosea, Rowell syndrome, and lichen planus have been reported following the Pfizer-BioNTech COVID-19 vaccine.1 Cutaneous reactions reported in association with the Sinovac-Coronavac COVID-19 vaccine include swelling, redness, itching, discoloration, induration (1%–10%), urticaria, petechial rash, and exacerbation of psoriasis at the local injection site (<1%).2

We describe 7 patients from Turkey who presented with various dermatologic problems 5 to 28 days after COVID-19 vaccination, highlighting the possibility of early and late cutaneous reactions related to the vaccine (Table).

CT11302016_e_Table.jpg

Case Reports

Patient 1—A 44-year-old woman was admitted to the dermatology clinic with painful lesions on the trunk of 3 days’ duration. Dermatologic examination revealed grouped erythematous vesicles showing dermatomal spread in the right thoracolumbar (dermatome T10) region. The patient reported that she had received 2 doses of the Sinovac-Coronavac vaccine (doses 1 and 2) and 2 doses of the BioNTech COVID-19 vaccine (doses 3 and 4); the rash had developed 28 days after she received the 4th dose. Her medical history was unremarkable. The lesions regressed after 1 week of treatment with oral valacyclovir 1000 mg 3 times daily, but she developed postherpetic neuralgia 1 week after starting treatment, which resolved after 8 weeks.

Patient 2—A 68-year-old woman presented to the dermatology clinic for evaluation of painful sores on the upper lip of 1 day’s duration. She had a history of rheumatoid arthritis, hypertension, and atopy and was currently taking prednisone and etanercept. Dermatologic examination revealed grouped vesicles on an erythematous base on the upper lip. A diagnosis of herpes labialis was made. The patient reported that she had received a third dose of the Sinovac-Coronavac vaccine 10 days prior to the appearance of the lesions. Her symptoms resolved completely within 2 weeks of treatment with topical acyclovir.

Patient 3—A 64-year-old woman was admitted to the hospital with pain, redness, and watery sores on and around the left eyelid of 2 days’ duration. Dermatologic evaluation revealed the erythematous surface of the left eyelid and periorbital area showed partial crusts, clustered vesicles, erythema, and edema. Additionally, the conjunctiva was purulent and erythematous. The patient’s medical history was notable for allergic asthma, hypertension, anxiety, and depression. For this reason, the patient was prescribed an angiotensin receptor blocker and a selective serotonin reuptake inhibitor. She noted that a similar rash had developed around the left eye 6 years prior that was diagnosed as herpes zoster (HZ). She also reported that she had received 2 doses of the Sinovac-Coronavac COVID-19 vaccine followed by 1 dose of the BioNTech COVID-19 vaccine, which she had received 2 weeks before the rash developed. The patient was treated at the eye clinic and was found to have ocular involvement. Ophthalmology was consulted and a diagnosis of herpes zoster ophthalmicus (HZO) was made. Systemic valacyclovir treatment was initiated, resulting in clinical improvement within 3 weeks.

Patient 4—A 75-year-old man was admitted to the hospital with chest and back pain and widespread muscle pain of several days’ duration. His medical history was remarkable for diabetes mellitus, hypertension, depression, and coronary artery bypass surgery. A medication history revealed treatment with a β-blocker, acetylsalicylic acid, a calcium channel blocker, a dipeptidyl peptidase 4 inhibitor, and a selective serotonin reuptake inhibitor. Dermatologic examination revealed grouped vesicles on an erythematous background in dermatome T5 on the right chest and back. A diagnosis of HZ was made. The patient reported that he had received 2 doses of the Sinovac-Coronavac vaccine followed by 1 dose of the Pfizer-BioNTech vaccine 2 weeks prior to the current presentation. He was treated with valacyclovir for 1 week, and his symptoms resolved entirely within 3 weeks.

Patient 5—A 50-year-old woman presented to the hospital for evaluation of painful sores on the back, chest, groin, and abdomen of 10 days’ duration. The lesions initially had developed 7 days after receiving the BioNTech COVID-19 vaccine; she previously had received 2 doses of the Sinovac-Coronavac vaccine. The patient had a history of untreated psoriasis. Dermatologic examination revealed grouped vesicles on an erythematous background in the T2–L2 dermatomes on the left side of the trunk. A diagnosis of HZ was made. The lesions resolved after 1 week of treatment with systemic valacyclovir; however, she subsequently developed postherpetic neuralgia, hypoesthesia, and postinflammatory hyperpigmentation in the affected regions.

 

 

Patient 6—A 37-year-old woman presented to the hospital with redness, swelling, and itching all over the body of 3 days’ duration. The patient noted that the rash would subside and reappear throughout the day. Her medical history was unremarkable, except for COVID-19 infection 6 months prior. She had received a second dose of the BioNTech vaccine 20 days prior to development of symptoms. Dermatologic examination revealed widespread erythematous urticarial plaques. A diagnosis of acute urticaria was made. The patient recovered completely after 1 week of treatment with a systemic steroid and 3 weeks of antihistamine treatment.

Patient 7—A 63-year-old woman presented to the hospital with widespread itching and rash that appeared 5 days after the first dose of the BioNTech COVID-19 vaccine. The patient reported that the rash resolved spontaneously within a few hours but then reappeared. Her medical history revealed that she was taking tamoxifen for breast cancer and that she previously had received 2 doses of the Sinovac-Coronavac vaccine. Dermatologic examination revealed erythematous urticarial plaques on the trunk and arms. A diagnosis of urticaria was made, and her symptoms resolved after 6 weeks of antihistamine treatment.

Comment

Skin lesions associated with COVID-19 infection have been reported worldwide3,4 as well as dermatologic reactions following COVID-19 vaccination. In one case from Turkey, HZ infection was reported in a 68-year-old man 5 days after he received a second dose of the COVID-19 vaccine.5 In another case, HZ infection developed in a 78-year-old man 5 days after COVID-19 vaccination.6 Numerous cases of HZ infection developing within 1 to 26 days of COVID-19 vaccination have been reported worldwide.7-9

In a study conducted in the United States, 40 skin reactions associated with the COVID-19 vaccine were investigated; of these cases, 87.5% (35/40) were reported as varicella-zoster virus, and 12.5% (5/40) were reported as herpes simplex reactivation; 54% (19/35) and 80% (4/5) of these cases, respectively, were associated with the Pfizer-BioNTech vaccine.10 The average age of patients who developed a skin reaction was 46 years, and 70% (28/40) were women. The time to onset of the reaction was 2 to 13 days after vaccination, and symptoms were reported to improve within 7 days on average.10

Another study from Spain examined 405 vaccine-related skin reactions, 40.2% of which were related to the Pfizer-BioNTech vaccine. Among them, 80.2% occurred in women; 13.8% of cases were diagnosed as varicella-zoster virus or HZ virus reactivation, and 14.6% were urticaria. Eighty reactions (21%) were classified as severe/very severe and 81% required treatment.11 One study reported 414 skin reactions from the COVID-19 vaccine from December 2020 to February 2021; of these cases, 83% occurred after the Moderna vaccine, which is not available in Turkey, and 17% occurred after the Pfizer-BioNTech vaccine.12A systematic review of 91 patients who developed HZ infection after COVID-19 vaccination reported that 10% (9/91) of cases were receiving immunosuppressive therapy and 13% (12/91) had an autoimmune disease.7 In our case series, it is known that at least 2 of the patients (patients 2 and 5), including 1 patient with rheumatoid arthritis (patient 2) who was on immunosuppressive treatment, had autoimmune disorders. However, reports in the literature indicate that most patients with autoimmune inflammatory rheumatic diseases remain stable after vaccination.13

Herpes zoster ophthalmicus is a rare form of HZ caused by involvement of the ophthalmic branch of the trigeminal nerve that manifests as vesicular lesions and retinitis, uveitis, keratitis, conjunctivitis, and pain on an erythematous background. Two cases of women who developed HZO infection after Pfizer-BioNTech vaccination were reported in the literature.14 Although patient 3 in our case series had a history of HZO 6 years prior, the possibility of the Pfizer-BioNTech vaccine triggering HZO should be taken into consideration.

Although cutaneous reactions after the Sinovac-Coronavac vaccine were observed in only 1 of 7 patients in our case series, skin reactions after Sinovac-Coronavac (an inactivated viral vaccine) have been reported in the literature. In one study, after a total of 35,229 injections, the incidence of cutaneous adverse events due to Sinovac-Coronavac was reported to be 0.94% and 0.70% after the first and second doses, respectively.15 Therefore, further study results are needed to directly attribute the reactions to COVID-19 vaccination.

Conclusion

Our case series highlights that clinicians should be vigilant in diagnosing cutaneous reactions following COVID-19 vaccination early to prevent potential complications. Early recognition of reactions is crucial, and the prognosis can be improved with appropriate treatment. Despite the potential dermatologic adverse effects of the COVID-19 vaccine, the most effective way to protect against serious COVID-19 infection is to continue to be vaccinated.

References
  1. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N Engl J Med. 2020;383:2603-2615.
  2. Zhang Y, Zeng G, Pan H, et al. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18–59 years: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect Dis. 2021;21:181-192.
  3. Tan SW, Tam YC, Oh CC. Skin manifestations of COVID-19: a worldwide review. JAAD Int. 2021;2:119-133.
  4. Singh H, Kaur H, Singh K, et al. Cutaneous manifestations of COVID-19: a systematic review. advances in wound care. 2021;10:51-80.
  5. Aksu SB, Öztürk GZ. A rare case of shingles after COVID-19 vaccine: is it a possible adverse effect? clinical and experimental vaccine research. 2021;10:198-201.
  6. Bostan E, Yalici-Armagan B. Herpes zoster following inactivated COVID-19 vaccine: a coexistence or coincidence? J Cosmet Dermatol. 2021;20:1566-1567.
  7. Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. Vaccines (Basel). 2021;9:1013. doi:10.3390/vaccines9091013
  8. Rodríguez-Jiménez P, Chicharro P, Cabrera LM, et al. Varicella-zoster virus reactivation after SARS-CoV-2 BNT162b2 mRNA vaccination: report of 5 cases. JAAD Case Rep. 2021;12:58-59. doi:10.1016/j.jdcr.2021.04.014
  9. Lee C, Cotter D, Basa J, et al. 20 Post-COVID-19 vaccine-related shingles cases seen at the Las Vegas Dermatology clinic and sent to us via social media. J Cosmet Dermatol. 2021;20:1960-1964.
  10. Fathy RA, McMahon DE, Lee C, et al. Varicella-zoster and herpes simplex virus reactivation post-COVID-19 vaccination: a review of 40 cases in an International Dermatology Registry. J Eur Acad Dermatol Venerol. 2022;36:E6-E9.
  11. Català A, Muñoz-Santos C, Galván-Casas C, et al. Cutaneous reactions after SARS-CoV-2 vaccination: a cross-sectional Spanish nationwide study of 405 cases. Br J Dermatol. 2022;186:142-152.
  12. McMahon DE, Amerson E, Rosenbach M, et al. Cutaneous reactions reported after Moderna and Pfizer COVID-19 vaccination: a registry-based study of 414 cases. J Am Acad Dermatol. 2021;85:46-55.
  13. Furer V, Eviatar T, Zisman D, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. Ann Rheum Dis. 2021;80:1330-1338.
  14. Bernardini N, Skroza N, Mambrin A, et al. Herpes zoster ophthalmicus in two women after Pfizer-BioNTech (BNT162b2) vaccine. J Med Virol. 2022;94:817-818.
  15. Rerknimitr P, Puaratanaarunkon T, Wongtada C, et al. Cutaneous adverse reactions from 35,229 doses of Sinovac and AstraZeneca COVID-19 vaccination: a prospective cohort study in healthcare workers. J Eur Acad Dermatol Venereol. 2022;36:E158-E161.
References
  1. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. N Engl J Med. 2020;383:2603-2615.
  2. Zhang Y, Zeng G, Pan H, et al. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18–59 years: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. Lancet Infect Dis. 2021;21:181-192.
  3. Tan SW, Tam YC, Oh CC. Skin manifestations of COVID-19: a worldwide review. JAAD Int. 2021;2:119-133.
  4. Singh H, Kaur H, Singh K, et al. Cutaneous manifestations of COVID-19: a systematic review. advances in wound care. 2021;10:51-80.
  5. Aksu SB, Öztürk GZ. A rare case of shingles after COVID-19 vaccine: is it a possible adverse effect? clinical and experimental vaccine research. 2021;10:198-201.
  6. Bostan E, Yalici-Armagan B. Herpes zoster following inactivated COVID-19 vaccine: a coexistence or coincidence? J Cosmet Dermatol. 2021;20:1566-1567.
  7. Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. Vaccines (Basel). 2021;9:1013. doi:10.3390/vaccines9091013
  8. Rodríguez-Jiménez P, Chicharro P, Cabrera LM, et al. Varicella-zoster virus reactivation after SARS-CoV-2 BNT162b2 mRNA vaccination: report of 5 cases. JAAD Case Rep. 2021;12:58-59. doi:10.1016/j.jdcr.2021.04.014
  9. Lee C, Cotter D, Basa J, et al. 20 Post-COVID-19 vaccine-related shingles cases seen at the Las Vegas Dermatology clinic and sent to us via social media. J Cosmet Dermatol. 2021;20:1960-1964.
  10. Fathy RA, McMahon DE, Lee C, et al. Varicella-zoster and herpes simplex virus reactivation post-COVID-19 vaccination: a review of 40 cases in an International Dermatology Registry. J Eur Acad Dermatol Venerol. 2022;36:E6-E9.
  11. Català A, Muñoz-Santos C, Galván-Casas C, et al. Cutaneous reactions after SARS-CoV-2 vaccination: a cross-sectional Spanish nationwide study of 405 cases. Br J Dermatol. 2022;186:142-152.
  12. McMahon DE, Amerson E, Rosenbach M, et al. Cutaneous reactions reported after Moderna and Pfizer COVID-19 vaccination: a registry-based study of 414 cases. J Am Acad Dermatol. 2021;85:46-55.
  13. Furer V, Eviatar T, Zisman D, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. Ann Rheum Dis. 2021;80:1330-1338.
  14. Bernardini N, Skroza N, Mambrin A, et al. Herpes zoster ophthalmicus in two women after Pfizer-BioNTech (BNT162b2) vaccine. J Med Virol. 2022;94:817-818.
  15. Rerknimitr P, Puaratanaarunkon T, Wongtada C, et al. Cutaneous adverse reactions from 35,229 doses of Sinovac and AstraZeneca COVID-19 vaccination: a prospective cohort study in healthcare workers. J Eur Acad Dermatol Venereol. 2022;36:E158-E161.
Issue
Cutis - 113(2)
Issue
Cutis - 113(2)
Page Number
E16-E19
Page Number
E16-E19
Publications
Publications
Topics
Article Type
Display Headline
Dermatologic Reactions Following COVID-19 Vaccination: A Case Series
Display Headline
Dermatologic Reactions Following COVID-19 Vaccination: A Case Series
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>Alkanat</fileName> <TBEID>0C02F2C1.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F2C1</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Alkanat</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240222T085847</firstPublished> <LastPublished>20240222T085848</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240222T085847</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Hafize Özdemir Alkanat, RN, PhD; Sevgi Kulaklý, MD</byline> <bylineText>Hafize Özdemir Alkanat, RN, PhD; Sevgi Kulaklı, MD</bylineText> <bylineFull>Hafize Özdemir Alkanat, RN, PhD; Sevgi Kulaklý, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E16-E19</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Cutaneous reactions associated with the Pfizer-BioNTech COVID-19 vaccine have been reported worldwide since December 2020. Local injection site reactions (&lt;1%) </metaDescription> <articlePDF>300329</articlePDF> <teaserImage/> <title>Dermatologic Reactions Following COVID-19 Vaccination: A Case Series</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>2165</CMSID> </CMSIDs> <keywords> <keyword>COVID-19</keyword> <keyword> infectious disease</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>February 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">45</term> </sections> <topics> <term>234</term> <term canonical="true">63993</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180026ce.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Dermatologic Reactions Following COVID-19 Vaccination: A Case Series</title> <deck/> </itemMeta> <itemContent> <p class="abstract">We describe 7 patients with various dermatologic reactions following COVID-19 vaccination, including herpes zoster (HZ) infection, herpes zoster ophthalmicus (HZO), herpes labialis, and urticaria. Although the reactions described here may be related to COVID-19 vaccination, continued vaccination is recommended, as it is the most effective way to protect against serious COVID-19 infection.</p> <p> <em><em>Cutis. </em>2024;113:E16-E19.</em> </p> <p>Cutaneous reactions associated with the Pfizer-BioNTech COVID-19 vaccine have been reported worldwide since December 2020. Local injection site reactions (<span class="body">&lt;</span>1%) such as erythema, swelling, delayed local reactions (1%–10%), morbilliform rash, urticarial reactions, pityriasis rosea, Rowell syndrome, and lichen planus have been reported following the Pfizer-BioNTech COVID-19 vaccine.<sup>1</sup> Cutaneous reactions reported in association with the Sinovac-Coronavac COVID-19 vaccine include swelling, redness, itching, discoloration, induration (1%–10%), urticaria, petechial rash, and exacerbation of psoriasis at the local injection site (<span class="body">&lt;</span>1%).<sup>2</sup></p> <p>We describe 7 patients from Turkey who presented with various dermatologic problems 5 to 28 days after COVID-19 vaccination, highlighting the possibility of early and late cutaneous reactions related to the vaccine (Table).</p> <h3>Case Reports</h3> <p><i>Patient 1—</i>A 44-year-old woman was admitted to the dermatology clinic with painful lesions on the trunk of 3 days’ duration. Dermatologic examination revealed grouped erythematous vesicles showing dermatomal spread in the right thoracolumbar (dermatome T10) region. The patient reported that she had received 2 doses of the Sinovac-Coronavac vaccine (doses 1 and 2) and 2 doses of the BioNTech COVID-19 vaccine (doses 3 and 4); the rash had developed 28 days after she received the 4th dose. Her medical history was unremarkable. The lesions regressed after 1 week of treatment with oral valacyclovir 1000 mg 3 times daily, but she developed postherpetic neuralgia 1 week after starting treatment, which resolved after 8 weeks.</p> <p><i>Patient 2—</i>A 68-year-old woman presented to the dermatology clinic for evaluation of painful sores on the upper lip of 1 day’s duration. She had a history of rheumatoid arthritis, hypertension, and atopy and was currently taking prednisone and etanercept. Dermatologic examination revealed grouped vesicles on an erythematous base on the upper lip. A diagnosis of herpes labialis was made. The patient reported that she had received a third dose of the Sinovac-Coronavac vaccine 10 days prior to the appearance of the lesions. Her symptoms resolved completely within 2 weeks of treatment with topical acyclovir.<br/><br/><i>Patient 3—</i>A 64-year-old woman was admitted to the hospital with pain, redness, and watery sores on and around the left eyelid of 2 days’ duration. Dermatologic evaluation revealed the erythematous surface of the left eyelid and periorbital area showed partial crusts, clustered vesicles, erythema, and edema. Additionally, the conjunctiva was purulent and erythematous.<span class="cf01"> </span>The patient’s medical history was notable for allergic asthma, hypertension, anxiety, and depression. For this reason, the patient was prescribed an angiotensin receptor blocker and a selective serotonin reuptake inhibitor. She noted that a similar rash had developed around the left eye 6 years prior that was diagnosed as herpes zoster (HZ). She also reported that she had received 2 doses of the Sinovac-Coronavac COVID-19 vaccine followed by 1 dose of the BioNTech COVID-19 vaccine, which she had received 2 weeks before the rash developed. The patient was treated at the eye clinic and was found to have ocular involvement. Ophthalmology was consulted and a diagnosis of herpes zoster ophthalmicus (HZO) was made. Systemic valacyclovir treatment was initiated, resulting in clinical improvement within 3 weeks.<br/><br/><i>Patient 4—</i>A 75-year-old man was admitted to the hospital with chest and back pain and widespread muscle pain of several days’ duration. His medical history was remarkable for diabetes mellitus, hypertension, depression, and coronary artery bypass surgery. A medication history revealed treatment with a β-blocker, acetylsalicylic acid, a calcium channel blocker, a dipeptidyl peptidase 4 inhibitor, and a selective serotonin reuptake inhibitor. Dermatologic examination revealed grouped vesicles on an erythematous background in dermatome T5 on the right chest and back. A diagnosis of HZ was made. The patient reported that he had received 2 doses of the Sinovac-Coronavac vaccine followed by 1 dose of the Pfizer-BioNTech vaccine 2 weeks prior to the current presentation. He was treated with valacyclovir for 1 week, and his symptoms resolved entirely within 3 weeks.<br/><br/><i>Patient 5—</i>A 50-year-old woman presented to the hospital for evaluation of painful sores on the back, chest, groin, and abdomen of 10 days’ duration. The lesions initially had developed 7 days after receiving the BioNTech COVID-19 vaccine; she previously had received 2 doses of the Sinovac-Coronavac vaccine. The patient had a history of untreated psoriasis. Dermatologic examination revealed grouped vesicles on an erythematous background in the T2–L2 dermatomes on the left side of the trunk. A diagnosis of HZ was made. The lesions resolved after 1 week of treatment with systemic valacyclovir; however, she subsequently developed postherpetic neuralgia, hypoesthesia, and postinflammatory hyperpigmentation in the affected regions.<br/><br/><i>Patient 6—</i>A 37-year-old woman presented to the hospital with redness, swelling, and itching all over the body of 3 days’ duration. The patient noted that the rash would subside and reappear throughout the day. Her medical history was unremarkable, except for COVID-19 infection 6 months prior. She had received a second dose of the BioNTech vaccine 20 days prior to development of symptoms. Dermatologic examination revealed widespread erythematous urticarial plaques. A diagnosis of acute urticaria was made. The patient recovered completely after 1 week of treatment with a systemic steroid and 3 weeks of antihistamine treatment.<br/><br/><i>Patient 7—</i>A 63-year-old woman presented to the hospital with widespread itching and rash that appeared 5 days after the first dose of the BioNTech COVID-19 vaccine. The patient reported that the rash resolved spontaneously within a few hours but then reappeared. Her medical history revealed that she was taking tamoxifen for breast cancer and that she previously had received 2 doses of the Sinovac-Coronavac vaccine. Dermatologic examination revealed erythematous urticarial plaques on the trunk and arms. A diagnosis of urticaria was made, and her symptoms resolved after 6 weeks of antihistamine treatment.</p> <h3>Comment</h3> <p>Skin lesions associated with COVID-19 infection have been reported worldwide<sup>3,4</sup> as well as dermatologic reactions following COVID-19 vaccination. In one case from Turkey, HZ infection was reported in a 68-year-old man 5 days after he received a second dose of the COVID-19 vaccine.<sup>5</sup> In another case, HZ infection developed in a 78-year-old man 5 days after COVID-19 vaccination.<sup>6</sup> Numerous cases of HZ infection developing within 1 to 26 days of COVID-19 vaccination have been reported worldwide.<sup>7-9</sup></p> <p>In a study conducted in the United States, 40 skin reactions associated with the COVID-19 vaccine were investigated; of these cases, 87.5% (35/40) were reported as varicella-zoster virus, and 12.5% (5/40) were reported as herpes simplex reactivation; 54% (19/35) and 80% (4/5) of these cases, respectively, were associated with the Pfizer-BioNTech vaccine.<sup>10</sup> The average age of patients who developed a skin reaction was 46 years, and 70% (28/40) were women. The time to onset of the reaction was 2 to 13 days after vaccination, and symptoms were reported to improve within 7 days on average.<sup>10</sup> <br/><br/>Another study from Spain examined 405 vaccine-related skin reactions, 40.2% of which were related to the Pfizer-BioNTech vaccine. Among them, 80.2% occurred in women; 13.8% of cases were diagnosed as varicella-zoster virus or HZ virus reactivation, and 14.6% were urticaria. Eighty reactions (21%) were classified as severe/very severe and 81% required treatment.<sup>11</sup> One study reported 414 skin reactions from the COVID-19 vaccine from December 2020 to February 2021; of these cases, 83% occurred after the Moderna vaccine, which is not available in Turkey, and 17% occurred after the Pfizer-BioNTech vaccine.<sup>12</sup><span class="markedcontent">A systematic review of 91 patients who developed HZ infection after COVID-19 vaccination reported that 10% (9/91) of cases were receiving immunosuppressive therapy and 13% (12/91) had an autoimmune disease.</span><span class="markedcontent"><sup>7</sup></span><span class="markedcontent"> </span>In our case series, it is known that at least 2 of the patients (patients 2 and 5), including 1 patient with rheumatoid arthritis (patient 2) who was on immunosuppressive treatment, had autoimmune disorders.<span class="markedcontent"> However, reports in the literature indicate that most patients with autoimmune inflammatory rheumatic diseases remain stable after vaccination.</span><sup>13</sup><br/><br/>Herpes zoster ophthalmicus is a rare form of HZ caused by involvement of the ophthalmic branch of the trigeminal nerve that manifests as vesicular lesions and retinitis, uveitis, keratitis, conjunctivitis, and pain on an erythematous background. Two cases of women who developed HZO infection after Pfizer-BioNTech vaccination were reported in the literature.<sup>14</sup> Although patient 3 in our case series had a history of HZO 6 years prior, the possibility of the Pfizer-BioNTech vaccine triggering HZO should be taken into consideration.<br/><br/>Although cutaneous reactions after the Sinovac-Coronavac vaccine were observed in only 1 of 7 patients in our case series, skin reactions after Sinovac-Coronavac (an inactivated viral vaccine) have been reported in the literature. In one study, after a total of 35,229 injections, the incidence of cutaneous adverse events due to Sinovac-Coronavac was reported to be 0.94% and 0.70% after the first and second doses, respectively.<sup>15</sup> Therefore, further study results are needed to directly attribute the reactions to COVID-19 vaccination.</p> <h3> <span class="y2iqfc">Conclusion</span> </h3> <p>Our case series highlights that clinicians should be vigilant in diagnosing cutaneous reactions following COVID-19 vaccination early to prevent potential complications. Early recognition of reactions is crucial, and the prognosis can be improved with appropriate treatment. Despite the potential dermatologic adverse effects of the COVID-19 vaccine, the most effective way to protect against serious COVID-19 infection is to continue to be vaccinated.</p> <h2>References</h2> <p class="reference"> 1. Polack FP, Thomas SJ, Kitchin N, et al. Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine. <span class="html-italic"><i>N Engl J Med.</i></span> 2020;<span class="html-italic">383:</span>2603-2615.<br/><br/> 2. Zhang Y, Zeng G, Pan H, et al. Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine in healthy adults aged 18–59 years: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial. <span class="html-italic"><i>Lancet Infect Dis.</i></span> 2021;<span class="html-italic">21:</span>181-192. <br/><br/> 3. Tan SW, Tam YC, Oh CC. Skin manifestations of COVID-19: a worldwide review. <i>JAAD Int</i>. 2021;2:119-133.<br/><br/> 4. Singh H, Kaur H, Singh K, et al. Cutaneous manifestations of COVID-19: a systematic review. advances in wound care. 2021;10:51-80.<br/><br/> 5. Aksu SB, Öztürk GZ. A rare case of shingles after COVID-19 vaccine: is it a possible adverse effect? clinical and experimental vaccine research. 2021;10:198-201.<br/><br/> 6. Bostan E, Yalici-Armagan B. Herpes zoster following inactivated COVID-19 vaccine: a coexistence or coincidence?<i> J Cosmet Dermatol.</i> 2021;20:1566-1567.<br/><br/> 7. Katsikas Triantafyllidis K, Giannos P, Mian IT, et al. Varicella zoster virus reactivation following COVID-19 vaccination: a systematic review of case reports. <i>Vaccines (Basel).</i> 2021;9:1013. <span class="citation-doi">doi:10.3390/vaccines9091013<br/><br/></span> 8. Rodríguez-Jiménez P, Chicharro P, Cabrera LM, et al. Varicella-zoster virus reactivation after SARS-CoV-2 BNT162b2 mRNA vaccination: report of 5 cases. <i>JAAD Case Rep</i>. 2021;12:58-59. <span class="citation-doi">doi:10.1016/j.jdcr.2021.04.014<br/><br/></span> 9. Lee C, Cotter D, Basa J, et al. 20 Post-COVID-19 vaccine-related shingles cases seen at the Las Vegas Dermatology clinic and sent to us via social media. <i>J Cosmet Dermatol. </i>2021;20:1960-1964.<br/><br/>10. Fathy RA, McMahon DE, Lee C, et al. Varicella-zoster and herpes simplex virus reactivation post-COVID-19 vaccination: a review of 40 cases in an International Dermatology Registry. <i>J Eur Acad Dermatol Venerol</i>. 2022;36:E6-E9.<br/><br/>11. Català A, Muñoz-Santos C, Galván-Casas C, et al. Cutaneous reactions after SARS-CoV-2 vaccination: a cross-sectional Spanish nationwide study of 405 cases. <i>Br J Dermatol. </i>2022;186:142-152.<br/><br/>12. McMahon DE, Amerson E, Rosenbach M, et al. Cutaneous reactions reported after Moderna and Pfizer COVID-19 vaccination: a registry-based study of 414 cases. <i>J Am Acad Dermatol</i>. 2021;85:46-55.<br/><br/>13. Furer V, Eviatar T, Zisman D, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. <i>Ann Rheum Dis.</i> 2021;80:1330-1338.<br/><br/>14. Bernardini N, Skroza N, Mambrin A, et al. Herpes zoster ophthalmicus in two women after Pfizer-BioNTech (BNT162b2) vaccine. <i>J Med Virol.</i> 2022;94:817-818.<br/><br/>15. Rerknimitr P, Puaratanaarunkon T, Wongtada C, et al. Cutaneous adverse reactions from 35,229 doses of Sinovac and AstraZeneca COVID-19 vaccination: a prospective cohort study in healthcare workers. <i>J Eur Acad Dermatol Venereol. </i>2022;36:E158-E161.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From Giresun University, Turkey. Dr. Alkanat is from the Faculty of Health Sciences, and Dr. Kulaklı is from the Faculty of Medicine, Dermatology Clinic.</p> <p class="disclosure">The authors report no conflict of interest.<br/><br/>Correspondence: Hafize Özdemir Alkanat, RN, PhD, Giresun University, Faculty of Health Sciences, 28340 Piraziz/Giresun, Turkey (hafize.ozdemir@giresun.edu.tr).<br/><br/>doi:10.12788/cutis.0966</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>Cutaneous reactions have been reported following COVID-19 vaccination.</li> <li>Herpes infections and urticarial reactions can be associated with COVID-19 vaccination, regardless of the delay in onset between the injection and symptom development.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Cutaneous reactions have been reported following COVID-19 vaccination.
  • Herpes infections and urticarial reactions can be associated with COVID-19 vaccination, regardless of the delay in onset between the injection and symptom development.
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
180026CE.SIG
Disable zoom
Off

My Kidney Is Fine, Can’t You Cystatin C?

Article Type
Changed
Sat, 02/03/2024 - 22:34

Clinicians usually measure renal function by using surrogate markers because directly measuring glomerular filtration rate (GFR) is not routinely feasible in a clinical setting.1,2 Creatinine (Cr) and cystatin C (CysC) are the 2 main surrogate molecules used to estimate GFR.3

Creatine is a molecule nonenzymatically converted into Cr, weighing only 113 Da in skeletal muscles.4 It is then filtered at the glomeruli and secreted at the proximal tubules of the kidneys. However, serum Cr (sCr) levels are affected by several factors, including age, biological sex, liver function, diet, and muscle mass.5 Historically, sCr levels also are affected by race.5 In an early study of factors affecting accurate GFR, researchers reported that self-identified African American patients had a 16% higher GFR than those who did not when using Cr.6 Despite this, the inclusion of Cr on a basic metabolic panel has allowed automatic reporting of an estimated GFR using sCr (eGFRCr) to be readily available.7

table.png

In comparison to Cr, CysC is an endogenous protein weighing 13 kDa produced by all nucleated cells.8,9 CysC is filtered by the kidney at the glomeruli and completely reabsorbed and catabolized by epithelial cells at the proximal tubule.9 Since production is not dependent on skeletal muscle, there are fewer physiological impacts on serum concentration of CysC. Levels of CysC may be elevated by factors shown in the Table.

Estimating Glomerular Filtration Rates

Multiple equations were developed to mitigate the impact of extraneous factors on the accuracy of an eGFRCr. The first widely used equation that included a variable adjustment for race was the Modification of Diet in Renal Disease study, presented in 2006.10 The equation increased the accuracy of eGFRCr further by adjusting for sex and age. It was followed by the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation in 2009, which was more accurate at higher GFR levels.11

CysC was simultaneously studied as an alternative to Cr with multiple equation iterations shown to be viable in various populations as early as 2003.12-15 However, it was not until 2012 that an equation for the use of CysC was offered for widespread use as an alternative to Cr alongside further refinement of the CKD-EPI equation for Cr.16 A new formula was presented in 2021 to use both sCr and serum CysC levels to obtain a more accurate estimation of GFR.17 Research continues its effort to accurately estimate GFR for diagnosing kidney disease and assessing comorbidities relating to decreased kidney function.3

All historical equations attempted to mitigate the potential impact of race on sCr level when calculating eGFRCrby assigning a separate variable for African American patients. As an unintended adverse effect, these equations may have led to discrimination by having a different equation for African American patients.18 Moreover, these Cr-based equations remain less accurate in patients with varied muscle mass, such as older patients, bodybuilders, athletes, and individuals with varied extremes of daily protein intake.1,8,9,19Several medications can also directly affect Cr clearance, reducing its ability to act as a surrogate for kidney function.1In this case report, we discuss an African American patient with high muscle mass and protein intake who was initially diagnosed with kidney disease based on an elevated Cr and found to be misdiagnosed based on the use of CysC for a more accurate GFR estimation.

 

 

Case Presentation

figure.png

A 35-year-old African American man serving in the military and recently diagnosed with HIV was referred to a nephrology clinic for further evaluation of an acute elevation in sCr. Before treatment for HIV, a brief record review showed a baseline Cr of about 1.3 mg/dL, with an eGFRCr of 75 mL/min/1.73 m2.20 In the same month, the patient was prescribed bictegravir/emtricitabine/tenofovir alafenamide, an HIV drug with nephrotoxic potential.21 The patient's total viral load remained low, and CD4 count remained > 500 after initiation of the HIV treatment. He was in his normal state of health and had no known contributory history before his HIV diagnosis. Cr readings peaked at 1.83 mg/dL after starting the HIV treatment and remained elevated to 1.73 mg/dL over the next few months, corresponding to CKD stage 3A. Because bictegravir/emtricitabine/tenofovir alafenamide is cleared by the kidneys and has a nephrotoxic profile, the clinical care team considered dosage adjustment or a medication switch given his observed elevated eGFRCr based on the CKD-EPI 2021 equation for Cr alone. It was also noted that the patient had a similar Cr spike to 1.83 mg/dL in 2018 without any identifiable renal insult or symptoms (Figure).

Diagnostic Evaluation

The primary care team ordered a renal ultrasound and referred the patient to the nephrology clinic. The nephrologist ordered the following laboratory studies: urine microalbumin to Cr ratio, basic metabolic panel (BMP), comprehensive metabolic panel (CMP), urinalysis, urine protein, urine Cr, parathyroid hormone level, hemoglobin A1c, complement component 3/4 panels, antinuclear and antineutrophil cytoplasmic antibodies titers, glomerular basement membrane antibody titer, urine light chains, serum protein electrophoresis, κ/λ ratio, viral hepatitis panel, and rapid plasma reagin testing. Much of this laboratory evaluation served to rule out any secondary causes of kidney disease, including autoimmune disease, monoclonal or polyclonal gammopathies, diabetic nephropathy or glomerulosclerosis, and nephrotic or nephritic syndromes.

All laboratory studies returned within normal limits; no proteinuria was discovered on urinalysis, and no abnormalities were visualized on renal ultrasound. Bictegravir/emtricitabine/tenofovir alafenamide nephrotoxicity was highest among the differential diagnoses due to the timing of Cr elevation coinciding with the initiation of the medications. The patient's CysC level was 0.85 mg/dL with a calculated eGFRCys of 125 mL/min/1.73 m2. The calculated sCR and serum cystatin C (eGFRCr-Cys) using the new 2021 equation and when adjusting for body surface area placed his eGFR at 92 mL/min/1.73 m2.20

The patient’s eGFRCysreassured the care team that the patient’s renal function was not acutely or chronically impacted by bictegravir/emtricitabine/tenofovir alafenamide, resulting in avoidance of unnecessary dosage adjustment or discontinuation of the HIV treatment. The patient reported a chronic habit of protein and creatine supplementation and bodybuilding, which likely further compounded the discrepancy between eGFRCr and eGFRCys and explained his previous elevation in Cr in 2018.

Follow-up

The patient underwent serial monitoring that revealed a stable Cr and unremarkable eGFR, ruling out CKD. There has been no evidence of worsening kidney disease to date, and the patient remained on his initial HIV regimen.

 

 

Discussion

This case shows the importance of using CysC as an alternative or confirmatory marker compared with sCr to estimate GFR in patients with high muscle mass and/or high creatine intake, such as many in the US Department of Defense (DoD) and US Department of Veterans Affairs (VA) patient populations. In the presented case, recorded Cr levels climbed from baseline Cr with the initiation of bictegravir/emtricitabine/tenofovir alafenamide. This raised the concern that HIV treatment was leading to the development of kidney damage.22

Diagnosis of kidney disease as opposed to the normal decline of eGFR with age in individuals without intrinsic CKD requires GFR ≥ 60 mL/min/1.73 m2 with kidney damage (proteinuria or radiological abnormalities, etc) or GFR < 135 to 140 mL/min/1.73 m2minus the patient’s age in years.23 The patient’s Cr peak at 1.83 mg/dL in 2018 led to an inappropriate diagnosis of kidney disease stage 3a based on an eGFRCr (2021 equation) of 52 mL/min/1.73 m2 when not corrected for body surface area.20 However, using the new 2021 equation using both Cr and CysC, the patient’s eGFRCr-Cyswas 92 mL/min/1.73 m2 after a correction for body surface area.

The 2009 CKD-EPI recommended the calculation of eGFR based on SCr concentration using age, sex, and race while the 2021 CKD-EPI recommended the exclusion of race.3 Both equations are less accurate in African American patients, individuals taking medications that interfere with Cr secretion and assay, and patients taking creatine supplements, high daily protein intake, or with high muscle mass.7 These settings result in a decreased eGFRCr without corresponding eGFRCys changes. Using SCr and CysC together, the eGFRCr-Cys yields improved concordance to measured GFR across race groups compared to GFR estimation based on Cr alone, which can avoid unnecessary expensive diagnostic workup, inappropriate kidney disease diagnosis, incorrect dosing of drugs, and accurately represent the military readiness of patients. Interestingly, in African American patients with recently diagnosed HIV, CKD-EPI using both Cr and CysC without race inclusion led to only a 2.9% overestimation of GFR and was the only equation with no statistically significant bias compared with measured GFR.24

A March 2023 case involving an otherwise healthy 26-year-old male active-duty US Navy member with a history of excessive protein supplement intake and intense exercise < 24 hours before laboratory work was diagnosed with CKD after a measured Cr of 16 mg/dL and an eGFRCr of 4 mL/min/1.73 m2 without any other evidence of kidney disease. His CysC remained within normal limits, resulting in a normal eGFRCys of 121 mL/min/1.73 m2, indicating no CKD. His Cr and eGFR recovered 10 days after his clinic visit and cessation of his supplement intake. These findings may not be uncommon given that 65% of active-duty military use protein supplements and 38% use other performance-enhancing supplements, such as creatine, according to a study.25

Unfortunately, the BMP/CMP traditionally used at VA centers use the eGFRCr equation, and it is unknown how many primary care practitioners recognize the limitations of these metabolic panels on accurate estimation of kidney function. However, in 2022 an expert panel including VA physicians recommended the immediate use of eGFRCr-Cys or eGFRCys for confirmatory testing and potentially screening of CKD.26 A small number of VAs have since adopted this recommendation, which should lead to fewer misdiagnoses among US military members as clinicians should now have access to more accurate measurements of GFR.

The VA spends about $18 billion (excluding dialysis) for care for 1.1 to 2.5 million VA patients with CKD.27 The majority of these diagnoses were undoubtedly made using the eGFRCr equation, raising the question of how many may be misdiagnosed. Assessment with CysC is currently relatively expensive, but it will likely become more affordable as the use of CysC as a confirmatory test increases.5 The cost of a sCr test is about $2.50, while CysC costs about $10.60, with variation from laboratory to laboratory.28 By comparison, a renal ultrasound costs $99 to $140 for uninsured patients.29 Furthermore, the cost of CysC testing is likely to trend downward as more facilities adopt the use of CysC measurements, which can be run on the same analytical equipment currently used for Cr measurements. Currently, most laboratories do not have established assays to use in-house and thus require CysC to be sent out to a laboratory, which increases result time and makes Cr a more attractive option. As more laboratories adopt assays for CysC, the cost of reagents will further decrease.

Given such considerations, confirmation testing of kidney function with CysC in specific patient populations with decreased eGFRCr without other features of CKD can offer great medical and financial benefits. A 2023 KDIGO report noted that many individuals may be mistakenly diagnosed with CKD when using eGFRCr.3 KDIGO noted that a 2013 meta-analysis of 90,000 individuals found that with a Cr-based eGFR of 45 to 59 mL/min/1.73 m2 (42%) had a CysC-based eGFR of ≥ 60 mL/min/1.73 m2. An eGFRCr of 45 to 59 represents 54% of all patients with CKD, amounting to millions of people (including current and former military personnel).3,29-31 Correcting a misdiagnosis of CKD would bring significant relief to patients and save millions in health care spending.

Conclusions

In patients who meet CKD criteria using eGFRCr but without other features of CKD, we recommend using confirmatory CysC levels and the eGFRCr-Cys equation. This will align care with the KDIGO guidelines and could be a cost-effective step toward improving military patient care. Further work in this area should focus on determining the knowledge gaps in primary care practitioners’ understanding of the limits of eGFRCr, the potential mitigation of concomitant CysC testing in equivocal CKD cases, and the cost-effectiveness and increased utilization of CysC.

References

1. Gabriel R. Time to scrap creatinine clearance? Br Med J (Clin Res Ed). 1986;293(6555):1119-1120. doi:10.1136/bmj.293.6555.1119

2. Swan SK. The search continues—an ideal marker of GFR. Clin Chem. 1997;43(6):913-914.doi:10.1093/clinchem/43.6.913 3. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013;3(1).

4. Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabolism. Physiol Rev. 2000;80(3):1107-1213. doi:10.1152/physrev.2000.80.3.1107

5. Ferguson TW, Komenda P, Tangri N. Cystatin C as a biomarker for estimating glomerular filtration rate. Curr Opin Nephrol Hypertens. 2015;24(3):295-300. doi:10.1097/mnh.0000000000000115

6. Levey AS, Titan SM, Powe NR, Coresh J, Inker LA. Kidney disease, race, and GFR estimation. Clin J Am Soc Nephrol. 2020;15(8):1203-1212. doi:10.2215/cjn.12791019

7. Shlipak MG, Tummalapalli SL, Boulware LE, et al; Conference Participants. The case for early identification and intervention of chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) controversies conference. Kidney Int. 2021;99(1):34-47. doi:10.1016/j.kint.2020.10.012

8. O’Riordan SE, Webb MC, Stowe HJ, et al. Cystatin C improves the detection of mild renal dysfunction in older patients. Ann Clin Biochem. 2003;40(pt 6):648-655. doi:10.1258/000456303770367243

9. Stevens LA, Schmid CH, Greene T, et al. Factors other than glomerular filtration rate affect serum cystatin C levels. Kidney Int. 2009;75(6):652-660. doi:10.1038/ki.2008.638

10. Levey AS, Coresh J, Greene T, et al; Chronic Kidney Disease Epidemiology Collaboration. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247-254. doi:10.7326/0003-4819-145-4-200608150-00004

11. Levey AS, Stevens LA, Schmid CH, et al; Chronic Kidney Disease Epidemiology Collaboration. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604-612. doi:10.7326/0003-4819-150-9-200905050-00006

12. Pöge U, Gerhardt T, Stoffel-Wagner B, Klehr HU, Sauerbruch T, Woitas RP. Calculation of glomerular filtration rate based on cystatin C in cirrhotic patients. Nephrol Dial Transplant. 2006;21(3):660-664. doi:10.1093/ndt/gfi305

13. Larsson A, Malm J, Grubb A, Hansson LO. Calculation of glomerular filtration rate expressed in mL/min from plasma cystatin C values in mg/L. Scand J Clin Lab Invest. 2004;64(1):25-30. doi:10.1080/00365510410003723.

14. Macisaac RJ, Tsalamandris C, Thomas MC, et al. Estimating glomerular filtration rate in diabetes: a comparison of cystatin-C- and creatinine-based methods. Diabetologia. 2006;49(7):1686-1689. doi:10.1007/s00125-006-0275-7

15. Rule AD, Bergstralh EJ, Slezak JM, Bergert J, Larson TS. Glomerular filtration rate estimated by cystatin C among different clinical presentations. Kidney Int. 2006;69(2):399-405. doi:10.1038/sj.ki.5000073

16. Inker LA, Schmid CH, Tighiouart H, et al; Chronic Kidney Disease Epidemiology Collaboration Investigators. Estimating glomerular filtration rate from serum creatinine and cystatin C. N Engl J Med. 2012;367(1):20-29. doi:10.1056/NEJMoa1114248

17. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234

18. Inker LA, Eneanya ND, Coresh J, et al; Chronic Kidney Disease Epidemiology Collaboration. New creatinine- and cystatin C–Based equations to estimate GFR without race. N Engl J Med. 2021;385(19):1737-1749. doi:10.1056/NEJMoa2102953

19. Oterdoom LH, Gansevoort RT, Schouten JP, de Jong PE, Gans ROB, Bakker SJL. Urinary creatinine excretion, an indirect measure of muscle mass, is an independent predictor of cardiovascular disease and mortality in the general population. Atherosclerosis. 2009;207(2):534-540. doi.10.1016/j.atherosclerosis.2009.05.010

20. National Kidney Foundation Inc. eGFR calculator. Accessed October 20, 2023. https://www.kidney.org/professionals/kdoqi/gfr_calculator

21. Ueaphongsukkit T, Gatechompol S, Avihingsanon A, et al. Tenofovir alafenamide nephrotoxicity: a case report and literature review. AIDS Res Ther. 2021;18(1):53. doi:10.1186/s12981-021-00380-w

22. D’Agati V, Appel GB. Renal pathology of human immunodeficiency virus infection. Semin Nephrol. 1998;18(4):406-421.

23. Glassock RJ, Winearls C. Ageing and the glomerular filtration rate: truths and consequences. Trans Am Clin Climatol Assoc. 2009;120:419-428.

24. Seape T, Gounden V, van Deventer HE, Candy GP, George JA. Cystatin C- and creatinine-based equations in the assessment of renal function in HIV-positive patients prior to commencing highly active antiretroviral therapy. Ann Clin Biochem. 2016;53(pt 1):58-66. doi:10.1177/0004563215579695

25. Tobin TW, Thurlow JS, Yuan CM. A healthy active duty soldier with an elevated serum creatinine. Mil Med. 2023;188(3-4):e866-e869. doi:10.1093/milmed/usab163

26. Delgado C, Baweja M, Crews DC, et al. A unifying approach for GFR estimation: recommendations of the NKF-ASN Task Force on Reassessing the Inclusion of Race in Diagnosing Kidney Disease. Am J Kidney Dis. 2022;79(2):268-288.e1. doi:10.1053/j.ajkd.2021.08.003

27. Saran R, Pearson A, Tilea A, et al; VA-REINS Steering Committee; VA Advisory Board. Burden and cost of caring for us veterans with CKD: initial findings from the VA Renal Information System (VA-REINS). Am J Kidney Dis. 2021;77(3):397-405. doi:10.1053/j.ajkd.2020.07.013

28. Zoler ML. Nephrologists make the case for cystatin C-based eGFR. Accessed October 20, 2023. https://www.medscape.com/viewarticle/951335#vp_2

29. Versaw N. How much does an ultrasound cost? Updated February 2022. Accessed October 20, 2023. https://www.compare.com/health/healthcare-resources/how-much-does-an-ultrasound-cost

30. Levey AS, Coresh J. Chronic kidney disease. Lancet. 2012;379(9811):165-180. doi:10.1016/S0140-6736(11)60178-5

31. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234

Article PDF
Author and Disclosure Information

Capt Alexander Beckstead, MD, USAFa; H. Reed Holmes, MDb; Capt Vi Tran, MD, USAFa; Bhagwan Dass, MDc

Correspondence: Bhagwan Dass (bhagwan.dass@va.gov)

aFamily Medicine Residency, Eglin Air Force Base, Florida

bDepartment of Internal Medicine, University of Florida, Gainesville

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

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 US Government, or any of its agencies.

Ethics and consent

Verbal and written informed consent for publication was obtained from the patient. All identifying patient information has been removed to protect patient privacy.

Issue
Federal Practitioner - 41(2)a
Publications
Topics
Page Number
62
Sections
Author and Disclosure Information

Capt Alexander Beckstead, MD, USAFa; H. Reed Holmes, MDb; Capt Vi Tran, MD, USAFa; Bhagwan Dass, MDc

Correspondence: Bhagwan Dass (bhagwan.dass@va.gov)

aFamily Medicine Residency, Eglin Air Force Base, Florida

bDepartment of Internal Medicine, University of Florida, Gainesville

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

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 US Government, or any of its agencies.

Ethics and consent

Verbal and written informed consent for publication was obtained from the patient. All identifying patient information has been removed to protect patient privacy.

Author and Disclosure Information

Capt Alexander Beckstead, MD, USAFa; H. Reed Holmes, MDb; Capt Vi Tran, MD, USAFa; Bhagwan Dass, MDc

Correspondence: Bhagwan Dass (bhagwan.dass@va.gov)

aFamily Medicine Residency, Eglin Air Force Base, Florida

bDepartment of Internal Medicine, University of Florida, Gainesville

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

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 US Government, or any of its agencies.

Ethics and consent

Verbal and written informed consent for publication was obtained from the patient. All identifying patient information has been removed to protect patient privacy.

Article PDF
Article PDF

Clinicians usually measure renal function by using surrogate markers because directly measuring glomerular filtration rate (GFR) is not routinely feasible in a clinical setting.1,2 Creatinine (Cr) and cystatin C (CysC) are the 2 main surrogate molecules used to estimate GFR.3

Creatine is a molecule nonenzymatically converted into Cr, weighing only 113 Da in skeletal muscles.4 It is then filtered at the glomeruli and secreted at the proximal tubules of the kidneys. However, serum Cr (sCr) levels are affected by several factors, including age, biological sex, liver function, diet, and muscle mass.5 Historically, sCr levels also are affected by race.5 In an early study of factors affecting accurate GFR, researchers reported that self-identified African American patients had a 16% higher GFR than those who did not when using Cr.6 Despite this, the inclusion of Cr on a basic metabolic panel has allowed automatic reporting of an estimated GFR using sCr (eGFRCr) to be readily available.7

table.png

In comparison to Cr, CysC is an endogenous protein weighing 13 kDa produced by all nucleated cells.8,9 CysC is filtered by the kidney at the glomeruli and completely reabsorbed and catabolized by epithelial cells at the proximal tubule.9 Since production is not dependent on skeletal muscle, there are fewer physiological impacts on serum concentration of CysC. Levels of CysC may be elevated by factors shown in the Table.

Estimating Glomerular Filtration Rates

Multiple equations were developed to mitigate the impact of extraneous factors on the accuracy of an eGFRCr. The first widely used equation that included a variable adjustment for race was the Modification of Diet in Renal Disease study, presented in 2006.10 The equation increased the accuracy of eGFRCr further by adjusting for sex and age. It was followed by the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation in 2009, which was more accurate at higher GFR levels.11

CysC was simultaneously studied as an alternative to Cr with multiple equation iterations shown to be viable in various populations as early as 2003.12-15 However, it was not until 2012 that an equation for the use of CysC was offered for widespread use as an alternative to Cr alongside further refinement of the CKD-EPI equation for Cr.16 A new formula was presented in 2021 to use both sCr and serum CysC levels to obtain a more accurate estimation of GFR.17 Research continues its effort to accurately estimate GFR for diagnosing kidney disease and assessing comorbidities relating to decreased kidney function.3

All historical equations attempted to mitigate the potential impact of race on sCr level when calculating eGFRCrby assigning a separate variable for African American patients. As an unintended adverse effect, these equations may have led to discrimination by having a different equation for African American patients.18 Moreover, these Cr-based equations remain less accurate in patients with varied muscle mass, such as older patients, bodybuilders, athletes, and individuals with varied extremes of daily protein intake.1,8,9,19Several medications can also directly affect Cr clearance, reducing its ability to act as a surrogate for kidney function.1In this case report, we discuss an African American patient with high muscle mass and protein intake who was initially diagnosed with kidney disease based on an elevated Cr and found to be misdiagnosed based on the use of CysC for a more accurate GFR estimation.

 

 

Case Presentation

figure.png

A 35-year-old African American man serving in the military and recently diagnosed with HIV was referred to a nephrology clinic for further evaluation of an acute elevation in sCr. Before treatment for HIV, a brief record review showed a baseline Cr of about 1.3 mg/dL, with an eGFRCr of 75 mL/min/1.73 m2.20 In the same month, the patient was prescribed bictegravir/emtricitabine/tenofovir alafenamide, an HIV drug with nephrotoxic potential.21 The patient's total viral load remained low, and CD4 count remained > 500 after initiation of the HIV treatment. He was in his normal state of health and had no known contributory history before his HIV diagnosis. Cr readings peaked at 1.83 mg/dL after starting the HIV treatment and remained elevated to 1.73 mg/dL over the next few months, corresponding to CKD stage 3A. Because bictegravir/emtricitabine/tenofovir alafenamide is cleared by the kidneys and has a nephrotoxic profile, the clinical care team considered dosage adjustment or a medication switch given his observed elevated eGFRCr based on the CKD-EPI 2021 equation for Cr alone. It was also noted that the patient had a similar Cr spike to 1.83 mg/dL in 2018 without any identifiable renal insult or symptoms (Figure).

Diagnostic Evaluation

The primary care team ordered a renal ultrasound and referred the patient to the nephrology clinic. The nephrologist ordered the following laboratory studies: urine microalbumin to Cr ratio, basic metabolic panel (BMP), comprehensive metabolic panel (CMP), urinalysis, urine protein, urine Cr, parathyroid hormone level, hemoglobin A1c, complement component 3/4 panels, antinuclear and antineutrophil cytoplasmic antibodies titers, glomerular basement membrane antibody titer, urine light chains, serum protein electrophoresis, κ/λ ratio, viral hepatitis panel, and rapid plasma reagin testing. Much of this laboratory evaluation served to rule out any secondary causes of kidney disease, including autoimmune disease, monoclonal or polyclonal gammopathies, diabetic nephropathy or glomerulosclerosis, and nephrotic or nephritic syndromes.

All laboratory studies returned within normal limits; no proteinuria was discovered on urinalysis, and no abnormalities were visualized on renal ultrasound. Bictegravir/emtricitabine/tenofovir alafenamide nephrotoxicity was highest among the differential diagnoses due to the timing of Cr elevation coinciding with the initiation of the medications. The patient's CysC level was 0.85 mg/dL with a calculated eGFRCys of 125 mL/min/1.73 m2. The calculated sCR and serum cystatin C (eGFRCr-Cys) using the new 2021 equation and when adjusting for body surface area placed his eGFR at 92 mL/min/1.73 m2.20

The patient’s eGFRCysreassured the care team that the patient’s renal function was not acutely or chronically impacted by bictegravir/emtricitabine/tenofovir alafenamide, resulting in avoidance of unnecessary dosage adjustment or discontinuation of the HIV treatment. The patient reported a chronic habit of protein and creatine supplementation and bodybuilding, which likely further compounded the discrepancy between eGFRCr and eGFRCys and explained his previous elevation in Cr in 2018.

Follow-up

The patient underwent serial monitoring that revealed a stable Cr and unremarkable eGFR, ruling out CKD. There has been no evidence of worsening kidney disease to date, and the patient remained on his initial HIV regimen.

 

 

Discussion

This case shows the importance of using CysC as an alternative or confirmatory marker compared with sCr to estimate GFR in patients with high muscle mass and/or high creatine intake, such as many in the US Department of Defense (DoD) and US Department of Veterans Affairs (VA) patient populations. In the presented case, recorded Cr levels climbed from baseline Cr with the initiation of bictegravir/emtricitabine/tenofovir alafenamide. This raised the concern that HIV treatment was leading to the development of kidney damage.22

Diagnosis of kidney disease as opposed to the normal decline of eGFR with age in individuals without intrinsic CKD requires GFR ≥ 60 mL/min/1.73 m2 with kidney damage (proteinuria or radiological abnormalities, etc) or GFR < 135 to 140 mL/min/1.73 m2minus the patient’s age in years.23 The patient’s Cr peak at 1.83 mg/dL in 2018 led to an inappropriate diagnosis of kidney disease stage 3a based on an eGFRCr (2021 equation) of 52 mL/min/1.73 m2 when not corrected for body surface area.20 However, using the new 2021 equation using both Cr and CysC, the patient’s eGFRCr-Cyswas 92 mL/min/1.73 m2 after a correction for body surface area.

The 2009 CKD-EPI recommended the calculation of eGFR based on SCr concentration using age, sex, and race while the 2021 CKD-EPI recommended the exclusion of race.3 Both equations are less accurate in African American patients, individuals taking medications that interfere with Cr secretion and assay, and patients taking creatine supplements, high daily protein intake, or with high muscle mass.7 These settings result in a decreased eGFRCr without corresponding eGFRCys changes. Using SCr and CysC together, the eGFRCr-Cys yields improved concordance to measured GFR across race groups compared to GFR estimation based on Cr alone, which can avoid unnecessary expensive diagnostic workup, inappropriate kidney disease diagnosis, incorrect dosing of drugs, and accurately represent the military readiness of patients. Interestingly, in African American patients with recently diagnosed HIV, CKD-EPI using both Cr and CysC without race inclusion led to only a 2.9% overestimation of GFR and was the only equation with no statistically significant bias compared with measured GFR.24

A March 2023 case involving an otherwise healthy 26-year-old male active-duty US Navy member with a history of excessive protein supplement intake and intense exercise < 24 hours before laboratory work was diagnosed with CKD after a measured Cr of 16 mg/dL and an eGFRCr of 4 mL/min/1.73 m2 without any other evidence of kidney disease. His CysC remained within normal limits, resulting in a normal eGFRCys of 121 mL/min/1.73 m2, indicating no CKD. His Cr and eGFR recovered 10 days after his clinic visit and cessation of his supplement intake. These findings may not be uncommon given that 65% of active-duty military use protein supplements and 38% use other performance-enhancing supplements, such as creatine, according to a study.25

Unfortunately, the BMP/CMP traditionally used at VA centers use the eGFRCr equation, and it is unknown how many primary care practitioners recognize the limitations of these metabolic panels on accurate estimation of kidney function. However, in 2022 an expert panel including VA physicians recommended the immediate use of eGFRCr-Cys or eGFRCys for confirmatory testing and potentially screening of CKD.26 A small number of VAs have since adopted this recommendation, which should lead to fewer misdiagnoses among US military members as clinicians should now have access to more accurate measurements of GFR.

The VA spends about $18 billion (excluding dialysis) for care for 1.1 to 2.5 million VA patients with CKD.27 The majority of these diagnoses were undoubtedly made using the eGFRCr equation, raising the question of how many may be misdiagnosed. Assessment with CysC is currently relatively expensive, but it will likely become more affordable as the use of CysC as a confirmatory test increases.5 The cost of a sCr test is about $2.50, while CysC costs about $10.60, with variation from laboratory to laboratory.28 By comparison, a renal ultrasound costs $99 to $140 for uninsured patients.29 Furthermore, the cost of CysC testing is likely to trend downward as more facilities adopt the use of CysC measurements, which can be run on the same analytical equipment currently used for Cr measurements. Currently, most laboratories do not have established assays to use in-house and thus require CysC to be sent out to a laboratory, which increases result time and makes Cr a more attractive option. As more laboratories adopt assays for CysC, the cost of reagents will further decrease.

Given such considerations, confirmation testing of kidney function with CysC in specific patient populations with decreased eGFRCr without other features of CKD can offer great medical and financial benefits. A 2023 KDIGO report noted that many individuals may be mistakenly diagnosed with CKD when using eGFRCr.3 KDIGO noted that a 2013 meta-analysis of 90,000 individuals found that with a Cr-based eGFR of 45 to 59 mL/min/1.73 m2 (42%) had a CysC-based eGFR of ≥ 60 mL/min/1.73 m2. An eGFRCr of 45 to 59 represents 54% of all patients with CKD, amounting to millions of people (including current and former military personnel).3,29-31 Correcting a misdiagnosis of CKD would bring significant relief to patients and save millions in health care spending.

Conclusions

In patients who meet CKD criteria using eGFRCr but without other features of CKD, we recommend using confirmatory CysC levels and the eGFRCr-Cys equation. This will align care with the KDIGO guidelines and could be a cost-effective step toward improving military patient care. Further work in this area should focus on determining the knowledge gaps in primary care practitioners’ understanding of the limits of eGFRCr, the potential mitigation of concomitant CysC testing in equivocal CKD cases, and the cost-effectiveness and increased utilization of CysC.

Clinicians usually measure renal function by using surrogate markers because directly measuring glomerular filtration rate (GFR) is not routinely feasible in a clinical setting.1,2 Creatinine (Cr) and cystatin C (CysC) are the 2 main surrogate molecules used to estimate GFR.3

Creatine is a molecule nonenzymatically converted into Cr, weighing only 113 Da in skeletal muscles.4 It is then filtered at the glomeruli and secreted at the proximal tubules of the kidneys. However, serum Cr (sCr) levels are affected by several factors, including age, biological sex, liver function, diet, and muscle mass.5 Historically, sCr levels also are affected by race.5 In an early study of factors affecting accurate GFR, researchers reported that self-identified African American patients had a 16% higher GFR than those who did not when using Cr.6 Despite this, the inclusion of Cr on a basic metabolic panel has allowed automatic reporting of an estimated GFR using sCr (eGFRCr) to be readily available.7

table.png

In comparison to Cr, CysC is an endogenous protein weighing 13 kDa produced by all nucleated cells.8,9 CysC is filtered by the kidney at the glomeruli and completely reabsorbed and catabolized by epithelial cells at the proximal tubule.9 Since production is not dependent on skeletal muscle, there are fewer physiological impacts on serum concentration of CysC. Levels of CysC may be elevated by factors shown in the Table.

Estimating Glomerular Filtration Rates

Multiple equations were developed to mitigate the impact of extraneous factors on the accuracy of an eGFRCr. The first widely used equation that included a variable adjustment for race was the Modification of Diet in Renal Disease study, presented in 2006.10 The equation increased the accuracy of eGFRCr further by adjusting for sex and age. It was followed by the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation in 2009, which was more accurate at higher GFR levels.11

CysC was simultaneously studied as an alternative to Cr with multiple equation iterations shown to be viable in various populations as early as 2003.12-15 However, it was not until 2012 that an equation for the use of CysC was offered for widespread use as an alternative to Cr alongside further refinement of the CKD-EPI equation for Cr.16 A new formula was presented in 2021 to use both sCr and serum CysC levels to obtain a more accurate estimation of GFR.17 Research continues its effort to accurately estimate GFR for diagnosing kidney disease and assessing comorbidities relating to decreased kidney function.3

All historical equations attempted to mitigate the potential impact of race on sCr level when calculating eGFRCrby assigning a separate variable for African American patients. As an unintended adverse effect, these equations may have led to discrimination by having a different equation for African American patients.18 Moreover, these Cr-based equations remain less accurate in patients with varied muscle mass, such as older patients, bodybuilders, athletes, and individuals with varied extremes of daily protein intake.1,8,9,19Several medications can also directly affect Cr clearance, reducing its ability to act as a surrogate for kidney function.1In this case report, we discuss an African American patient with high muscle mass and protein intake who was initially diagnosed with kidney disease based on an elevated Cr and found to be misdiagnosed based on the use of CysC for a more accurate GFR estimation.

 

 

Case Presentation

figure.png

A 35-year-old African American man serving in the military and recently diagnosed with HIV was referred to a nephrology clinic for further evaluation of an acute elevation in sCr. Before treatment for HIV, a brief record review showed a baseline Cr of about 1.3 mg/dL, with an eGFRCr of 75 mL/min/1.73 m2.20 In the same month, the patient was prescribed bictegravir/emtricitabine/tenofovir alafenamide, an HIV drug with nephrotoxic potential.21 The patient's total viral load remained low, and CD4 count remained > 500 after initiation of the HIV treatment. He was in his normal state of health and had no known contributory history before his HIV diagnosis. Cr readings peaked at 1.83 mg/dL after starting the HIV treatment and remained elevated to 1.73 mg/dL over the next few months, corresponding to CKD stage 3A. Because bictegravir/emtricitabine/tenofovir alafenamide is cleared by the kidneys and has a nephrotoxic profile, the clinical care team considered dosage adjustment or a medication switch given his observed elevated eGFRCr based on the CKD-EPI 2021 equation for Cr alone. It was also noted that the patient had a similar Cr spike to 1.83 mg/dL in 2018 without any identifiable renal insult or symptoms (Figure).

Diagnostic Evaluation

The primary care team ordered a renal ultrasound and referred the patient to the nephrology clinic. The nephrologist ordered the following laboratory studies: urine microalbumin to Cr ratio, basic metabolic panel (BMP), comprehensive metabolic panel (CMP), urinalysis, urine protein, urine Cr, parathyroid hormone level, hemoglobin A1c, complement component 3/4 panels, antinuclear and antineutrophil cytoplasmic antibodies titers, glomerular basement membrane antibody titer, urine light chains, serum protein electrophoresis, κ/λ ratio, viral hepatitis panel, and rapid plasma reagin testing. Much of this laboratory evaluation served to rule out any secondary causes of kidney disease, including autoimmune disease, monoclonal or polyclonal gammopathies, diabetic nephropathy or glomerulosclerosis, and nephrotic or nephritic syndromes.

All laboratory studies returned within normal limits; no proteinuria was discovered on urinalysis, and no abnormalities were visualized on renal ultrasound. Bictegravir/emtricitabine/tenofovir alafenamide nephrotoxicity was highest among the differential diagnoses due to the timing of Cr elevation coinciding with the initiation of the medications. The patient's CysC level was 0.85 mg/dL with a calculated eGFRCys of 125 mL/min/1.73 m2. The calculated sCR and serum cystatin C (eGFRCr-Cys) using the new 2021 equation and when adjusting for body surface area placed his eGFR at 92 mL/min/1.73 m2.20

The patient’s eGFRCysreassured the care team that the patient’s renal function was not acutely or chronically impacted by bictegravir/emtricitabine/tenofovir alafenamide, resulting in avoidance of unnecessary dosage adjustment or discontinuation of the HIV treatment. The patient reported a chronic habit of protein and creatine supplementation and bodybuilding, which likely further compounded the discrepancy between eGFRCr and eGFRCys and explained his previous elevation in Cr in 2018.

Follow-up

The patient underwent serial monitoring that revealed a stable Cr and unremarkable eGFR, ruling out CKD. There has been no evidence of worsening kidney disease to date, and the patient remained on his initial HIV regimen.

 

 

Discussion

This case shows the importance of using CysC as an alternative or confirmatory marker compared with sCr to estimate GFR in patients with high muscle mass and/or high creatine intake, such as many in the US Department of Defense (DoD) and US Department of Veterans Affairs (VA) patient populations. In the presented case, recorded Cr levels climbed from baseline Cr with the initiation of bictegravir/emtricitabine/tenofovir alafenamide. This raised the concern that HIV treatment was leading to the development of kidney damage.22

Diagnosis of kidney disease as opposed to the normal decline of eGFR with age in individuals without intrinsic CKD requires GFR ≥ 60 mL/min/1.73 m2 with kidney damage (proteinuria or radiological abnormalities, etc) or GFR < 135 to 140 mL/min/1.73 m2minus the patient’s age in years.23 The patient’s Cr peak at 1.83 mg/dL in 2018 led to an inappropriate diagnosis of kidney disease stage 3a based on an eGFRCr (2021 equation) of 52 mL/min/1.73 m2 when not corrected for body surface area.20 However, using the new 2021 equation using both Cr and CysC, the patient’s eGFRCr-Cyswas 92 mL/min/1.73 m2 after a correction for body surface area.

The 2009 CKD-EPI recommended the calculation of eGFR based on SCr concentration using age, sex, and race while the 2021 CKD-EPI recommended the exclusion of race.3 Both equations are less accurate in African American patients, individuals taking medications that interfere with Cr secretion and assay, and patients taking creatine supplements, high daily protein intake, or with high muscle mass.7 These settings result in a decreased eGFRCr without corresponding eGFRCys changes. Using SCr and CysC together, the eGFRCr-Cys yields improved concordance to measured GFR across race groups compared to GFR estimation based on Cr alone, which can avoid unnecessary expensive diagnostic workup, inappropriate kidney disease diagnosis, incorrect dosing of drugs, and accurately represent the military readiness of patients. Interestingly, in African American patients with recently diagnosed HIV, CKD-EPI using both Cr and CysC without race inclusion led to only a 2.9% overestimation of GFR and was the only equation with no statistically significant bias compared with measured GFR.24

A March 2023 case involving an otherwise healthy 26-year-old male active-duty US Navy member with a history of excessive protein supplement intake and intense exercise < 24 hours before laboratory work was diagnosed with CKD after a measured Cr of 16 mg/dL and an eGFRCr of 4 mL/min/1.73 m2 without any other evidence of kidney disease. His CysC remained within normal limits, resulting in a normal eGFRCys of 121 mL/min/1.73 m2, indicating no CKD. His Cr and eGFR recovered 10 days after his clinic visit and cessation of his supplement intake. These findings may not be uncommon given that 65% of active-duty military use protein supplements and 38% use other performance-enhancing supplements, such as creatine, according to a study.25

Unfortunately, the BMP/CMP traditionally used at VA centers use the eGFRCr equation, and it is unknown how many primary care practitioners recognize the limitations of these metabolic panels on accurate estimation of kidney function. However, in 2022 an expert panel including VA physicians recommended the immediate use of eGFRCr-Cys or eGFRCys for confirmatory testing and potentially screening of CKD.26 A small number of VAs have since adopted this recommendation, which should lead to fewer misdiagnoses among US military members as clinicians should now have access to more accurate measurements of GFR.

The VA spends about $18 billion (excluding dialysis) for care for 1.1 to 2.5 million VA patients with CKD.27 The majority of these diagnoses were undoubtedly made using the eGFRCr equation, raising the question of how many may be misdiagnosed. Assessment with CysC is currently relatively expensive, but it will likely become more affordable as the use of CysC as a confirmatory test increases.5 The cost of a sCr test is about $2.50, while CysC costs about $10.60, with variation from laboratory to laboratory.28 By comparison, a renal ultrasound costs $99 to $140 for uninsured patients.29 Furthermore, the cost of CysC testing is likely to trend downward as more facilities adopt the use of CysC measurements, which can be run on the same analytical equipment currently used for Cr measurements. Currently, most laboratories do not have established assays to use in-house and thus require CysC to be sent out to a laboratory, which increases result time and makes Cr a more attractive option. As more laboratories adopt assays for CysC, the cost of reagents will further decrease.

Given such considerations, confirmation testing of kidney function with CysC in specific patient populations with decreased eGFRCr without other features of CKD can offer great medical and financial benefits. A 2023 KDIGO report noted that many individuals may be mistakenly diagnosed with CKD when using eGFRCr.3 KDIGO noted that a 2013 meta-analysis of 90,000 individuals found that with a Cr-based eGFR of 45 to 59 mL/min/1.73 m2 (42%) had a CysC-based eGFR of ≥ 60 mL/min/1.73 m2. An eGFRCr of 45 to 59 represents 54% of all patients with CKD, amounting to millions of people (including current and former military personnel).3,29-31 Correcting a misdiagnosis of CKD would bring significant relief to patients and save millions in health care spending.

Conclusions

In patients who meet CKD criteria using eGFRCr but without other features of CKD, we recommend using confirmatory CysC levels and the eGFRCr-Cys equation. This will align care with the KDIGO guidelines and could be a cost-effective step toward improving military patient care. Further work in this area should focus on determining the knowledge gaps in primary care practitioners’ understanding of the limits of eGFRCr, the potential mitigation of concomitant CysC testing in equivocal CKD cases, and the cost-effectiveness and increased utilization of CysC.

References

1. Gabriel R. Time to scrap creatinine clearance? Br Med J (Clin Res Ed). 1986;293(6555):1119-1120. doi:10.1136/bmj.293.6555.1119

2. Swan SK. The search continues—an ideal marker of GFR. Clin Chem. 1997;43(6):913-914.doi:10.1093/clinchem/43.6.913 3. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013;3(1).

4. Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabolism. Physiol Rev. 2000;80(3):1107-1213. doi:10.1152/physrev.2000.80.3.1107

5. Ferguson TW, Komenda P, Tangri N. Cystatin C as a biomarker for estimating glomerular filtration rate. Curr Opin Nephrol Hypertens. 2015;24(3):295-300. doi:10.1097/mnh.0000000000000115

6. Levey AS, Titan SM, Powe NR, Coresh J, Inker LA. Kidney disease, race, and GFR estimation. Clin J Am Soc Nephrol. 2020;15(8):1203-1212. doi:10.2215/cjn.12791019

7. Shlipak MG, Tummalapalli SL, Boulware LE, et al; Conference Participants. The case for early identification and intervention of chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) controversies conference. Kidney Int. 2021;99(1):34-47. doi:10.1016/j.kint.2020.10.012

8. O’Riordan SE, Webb MC, Stowe HJ, et al. Cystatin C improves the detection of mild renal dysfunction in older patients. Ann Clin Biochem. 2003;40(pt 6):648-655. doi:10.1258/000456303770367243

9. Stevens LA, Schmid CH, Greene T, et al. Factors other than glomerular filtration rate affect serum cystatin C levels. Kidney Int. 2009;75(6):652-660. doi:10.1038/ki.2008.638

10. Levey AS, Coresh J, Greene T, et al; Chronic Kidney Disease Epidemiology Collaboration. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247-254. doi:10.7326/0003-4819-145-4-200608150-00004

11. Levey AS, Stevens LA, Schmid CH, et al; Chronic Kidney Disease Epidemiology Collaboration. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604-612. doi:10.7326/0003-4819-150-9-200905050-00006

12. Pöge U, Gerhardt T, Stoffel-Wagner B, Klehr HU, Sauerbruch T, Woitas RP. Calculation of glomerular filtration rate based on cystatin C in cirrhotic patients. Nephrol Dial Transplant. 2006;21(3):660-664. doi:10.1093/ndt/gfi305

13. Larsson A, Malm J, Grubb A, Hansson LO. Calculation of glomerular filtration rate expressed in mL/min from plasma cystatin C values in mg/L. Scand J Clin Lab Invest. 2004;64(1):25-30. doi:10.1080/00365510410003723.

14. Macisaac RJ, Tsalamandris C, Thomas MC, et al. Estimating glomerular filtration rate in diabetes: a comparison of cystatin-C- and creatinine-based methods. Diabetologia. 2006;49(7):1686-1689. doi:10.1007/s00125-006-0275-7

15. Rule AD, Bergstralh EJ, Slezak JM, Bergert J, Larson TS. Glomerular filtration rate estimated by cystatin C among different clinical presentations. Kidney Int. 2006;69(2):399-405. doi:10.1038/sj.ki.5000073

16. Inker LA, Schmid CH, Tighiouart H, et al; Chronic Kidney Disease Epidemiology Collaboration Investigators. Estimating glomerular filtration rate from serum creatinine and cystatin C. N Engl J Med. 2012;367(1):20-29. doi:10.1056/NEJMoa1114248

17. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234

18. Inker LA, Eneanya ND, Coresh J, et al; Chronic Kidney Disease Epidemiology Collaboration. New creatinine- and cystatin C–Based equations to estimate GFR without race. N Engl J Med. 2021;385(19):1737-1749. doi:10.1056/NEJMoa2102953

19. Oterdoom LH, Gansevoort RT, Schouten JP, de Jong PE, Gans ROB, Bakker SJL. Urinary creatinine excretion, an indirect measure of muscle mass, is an independent predictor of cardiovascular disease and mortality in the general population. Atherosclerosis. 2009;207(2):534-540. doi.10.1016/j.atherosclerosis.2009.05.010

20. National Kidney Foundation Inc. eGFR calculator. Accessed October 20, 2023. https://www.kidney.org/professionals/kdoqi/gfr_calculator

21. Ueaphongsukkit T, Gatechompol S, Avihingsanon A, et al. Tenofovir alafenamide nephrotoxicity: a case report and literature review. AIDS Res Ther. 2021;18(1):53. doi:10.1186/s12981-021-00380-w

22. D’Agati V, Appel GB. Renal pathology of human immunodeficiency virus infection. Semin Nephrol. 1998;18(4):406-421.

23. Glassock RJ, Winearls C. Ageing and the glomerular filtration rate: truths and consequences. Trans Am Clin Climatol Assoc. 2009;120:419-428.

24. Seape T, Gounden V, van Deventer HE, Candy GP, George JA. Cystatin C- and creatinine-based equations in the assessment of renal function in HIV-positive patients prior to commencing highly active antiretroviral therapy. Ann Clin Biochem. 2016;53(pt 1):58-66. doi:10.1177/0004563215579695

25. Tobin TW, Thurlow JS, Yuan CM. A healthy active duty soldier with an elevated serum creatinine. Mil Med. 2023;188(3-4):e866-e869. doi:10.1093/milmed/usab163

26. Delgado C, Baweja M, Crews DC, et al. A unifying approach for GFR estimation: recommendations of the NKF-ASN Task Force on Reassessing the Inclusion of Race in Diagnosing Kidney Disease. Am J Kidney Dis. 2022;79(2):268-288.e1. doi:10.1053/j.ajkd.2021.08.003

27. Saran R, Pearson A, Tilea A, et al; VA-REINS Steering Committee; VA Advisory Board. Burden and cost of caring for us veterans with CKD: initial findings from the VA Renal Information System (VA-REINS). Am J Kidney Dis. 2021;77(3):397-405. doi:10.1053/j.ajkd.2020.07.013

28. Zoler ML. Nephrologists make the case for cystatin C-based eGFR. Accessed October 20, 2023. https://www.medscape.com/viewarticle/951335#vp_2

29. Versaw N. How much does an ultrasound cost? Updated February 2022. Accessed October 20, 2023. https://www.compare.com/health/healthcare-resources/how-much-does-an-ultrasound-cost

30. Levey AS, Coresh J. Chronic kidney disease. Lancet. 2012;379(9811):165-180. doi:10.1016/S0140-6736(11)60178-5

31. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234

References

1. Gabriel R. Time to scrap creatinine clearance? Br Med J (Clin Res Ed). 1986;293(6555):1119-1120. doi:10.1136/bmj.293.6555.1119

2. Swan SK. The search continues—an ideal marker of GFR. Clin Chem. 1997;43(6):913-914.doi:10.1093/clinchem/43.6.913 3. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kidney Int Suppl. 2013;3(1).

4. Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabolism. Physiol Rev. 2000;80(3):1107-1213. doi:10.1152/physrev.2000.80.3.1107

5. Ferguson TW, Komenda P, Tangri N. Cystatin C as a biomarker for estimating glomerular filtration rate. Curr Opin Nephrol Hypertens. 2015;24(3):295-300. doi:10.1097/mnh.0000000000000115

6. Levey AS, Titan SM, Powe NR, Coresh J, Inker LA. Kidney disease, race, and GFR estimation. Clin J Am Soc Nephrol. 2020;15(8):1203-1212. doi:10.2215/cjn.12791019

7. Shlipak MG, Tummalapalli SL, Boulware LE, et al; Conference Participants. The case for early identification and intervention of chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) controversies conference. Kidney Int. 2021;99(1):34-47. doi:10.1016/j.kint.2020.10.012

8. O’Riordan SE, Webb MC, Stowe HJ, et al. Cystatin C improves the detection of mild renal dysfunction in older patients. Ann Clin Biochem. 2003;40(pt 6):648-655. doi:10.1258/000456303770367243

9. Stevens LA, Schmid CH, Greene T, et al. Factors other than glomerular filtration rate affect serum cystatin C levels. Kidney Int. 2009;75(6):652-660. doi:10.1038/ki.2008.638

10. Levey AS, Coresh J, Greene T, et al; Chronic Kidney Disease Epidemiology Collaboration. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247-254. doi:10.7326/0003-4819-145-4-200608150-00004

11. Levey AS, Stevens LA, Schmid CH, et al; Chronic Kidney Disease Epidemiology Collaboration. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604-612. doi:10.7326/0003-4819-150-9-200905050-00006

12. Pöge U, Gerhardt T, Stoffel-Wagner B, Klehr HU, Sauerbruch T, Woitas RP. Calculation of glomerular filtration rate based on cystatin C in cirrhotic patients. Nephrol Dial Transplant. 2006;21(3):660-664. doi:10.1093/ndt/gfi305

13. Larsson A, Malm J, Grubb A, Hansson LO. Calculation of glomerular filtration rate expressed in mL/min from plasma cystatin C values in mg/L. Scand J Clin Lab Invest. 2004;64(1):25-30. doi:10.1080/00365510410003723.

14. Macisaac RJ, Tsalamandris C, Thomas MC, et al. Estimating glomerular filtration rate in diabetes: a comparison of cystatin-C- and creatinine-based methods. Diabetologia. 2006;49(7):1686-1689. doi:10.1007/s00125-006-0275-7

15. Rule AD, Bergstralh EJ, Slezak JM, Bergert J, Larson TS. Glomerular filtration rate estimated by cystatin C among different clinical presentations. Kidney Int. 2006;69(2):399-405. doi:10.1038/sj.ki.5000073

16. Inker LA, Schmid CH, Tighiouart H, et al; Chronic Kidney Disease Epidemiology Collaboration Investigators. Estimating glomerular filtration rate from serum creatinine and cystatin C. N Engl J Med. 2012;367(1):20-29. doi:10.1056/NEJMoa1114248

17. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234

18. Inker LA, Eneanya ND, Coresh J, et al; Chronic Kidney Disease Epidemiology Collaboration. New creatinine- and cystatin C–Based equations to estimate GFR without race. N Engl J Med. 2021;385(19):1737-1749. doi:10.1056/NEJMoa2102953

19. Oterdoom LH, Gansevoort RT, Schouten JP, de Jong PE, Gans ROB, Bakker SJL. Urinary creatinine excretion, an indirect measure of muscle mass, is an independent predictor of cardiovascular disease and mortality in the general population. Atherosclerosis. 2009;207(2):534-540. doi.10.1016/j.atherosclerosis.2009.05.010

20. National Kidney Foundation Inc. eGFR calculator. Accessed October 20, 2023. https://www.kidney.org/professionals/kdoqi/gfr_calculator

21. Ueaphongsukkit T, Gatechompol S, Avihingsanon A, et al. Tenofovir alafenamide nephrotoxicity: a case report and literature review. AIDS Res Ther. 2021;18(1):53. doi:10.1186/s12981-021-00380-w

22. D’Agati V, Appel GB. Renal pathology of human immunodeficiency virus infection. Semin Nephrol. 1998;18(4):406-421.

23. Glassock RJ, Winearls C. Ageing and the glomerular filtration rate: truths and consequences. Trans Am Clin Climatol Assoc. 2009;120:419-428.

24. Seape T, Gounden V, van Deventer HE, Candy GP, George JA. Cystatin C- and creatinine-based equations in the assessment of renal function in HIV-positive patients prior to commencing highly active antiretroviral therapy. Ann Clin Biochem. 2016;53(pt 1):58-66. doi:10.1177/0004563215579695

25. Tobin TW, Thurlow JS, Yuan CM. A healthy active duty soldier with an elevated serum creatinine. Mil Med. 2023;188(3-4):e866-e869. doi:10.1093/milmed/usab163

26. Delgado C, Baweja M, Crews DC, et al. A unifying approach for GFR estimation: recommendations of the NKF-ASN Task Force on Reassessing the Inclusion of Race in Diagnosing Kidney Disease. Am J Kidney Dis. 2022;79(2):268-288.e1. doi:10.1053/j.ajkd.2021.08.003

27. Saran R, Pearson A, Tilea A, et al; VA-REINS Steering Committee; VA Advisory Board. Burden and cost of caring for us veterans with CKD: initial findings from the VA Renal Information System (VA-REINS). Am J Kidney Dis. 2021;77(3):397-405. doi:10.1053/j.ajkd.2020.07.013

28. Zoler ML. Nephrologists make the case for cystatin C-based eGFR. Accessed October 20, 2023. https://www.medscape.com/viewarticle/951335#vp_2

29. Versaw N. How much does an ultrasound cost? Updated February 2022. Accessed October 20, 2023. https://www.compare.com/health/healthcare-resources/how-much-does-an-ultrasound-cost

30. Levey AS, Coresh J. Chronic kidney disease. Lancet. 2012;379(9811):165-180. doi:10.1016/S0140-6736(11)60178-5

31. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. N Engl J Med. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234

Issue
Federal Practitioner - 41(2)a
Issue
Federal Practitioner - 41(2)a
Page Number
62
Page Number
62
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 Cystain</fileName> <TBEID>0C02EF62.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EF62</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240201T120113</firstPublished> <LastPublished>20240201T120113</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240201T120113</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Capt Alexander Beckstead, MD, USAFa; H. Reed Holmes, MDb; Capt Vi Tran, MD, USAFa; Bhagwan Dass, MDc</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>Clinicians usually measure renal function by using surrogate markers because directly measuring glomerular filtration rate (GFR) is not routinely feasible in a </metaDescription> <articlePDF/> <teaserImage/> <title>My Kidney Is Fine, Can’t You Cystatin C?</title> <deck/> <eyebrow>Case in Point</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>February</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>2</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2953</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>February 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case in Point | 2953<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">45</term> </sections> <topics> <term canonical="true">255</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>My Kidney Is Fine, Can’t You Cystatin C?</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b><b> </b>Independent of age, sex, and body composition, individuals of African American race and individuals with high muscle mass have elevated serum creatinine (sCr) levels on average that may result in overestimation of chronic kidney disease (CKD). We present a misdiagnosed case of CKD based on sCr levels, illustrating the utility of cystatin C (CysC) confirmation testing to answer the question: Can confirmation screening of kidney function with CysC in African American patients and patients with high muscle mass reduce the misdiagnosis of CKD?<br/><br/><b>Case Presentation: </b>A 35-year-old African American man with a history of well-controlled HIV was found to have consistently elevated creatinine (Cr). We diagnosed CKD stage 3A based on the estimated glomerular filtration rate (eGFR). Further evaluation showed isolated elevation of sCr with unremarkable urinalysis and other laboratory tests. sCr elevation predated diagnosis and HIV treatment. A CysC-based eGFR (eGFR<sub>cys</sub>) test confirmed the absence of CKD. <br/><br/><b>Conclusions: </b>The 2009 CKD Epidemiology Collaboration calculation of eGFR based on sCr concentration uses age, sex, and race, with an updated recommendation in 2021 to exclude race. Both equations are less accurate in African American patients, individuals taking medications that interfere with sCr secretion and assay, and patients taking creatine supplements or high protein intake. These clinical scenarios decrease sCr-based eGFR (eGFR<sub>Cr</sub>) but do not change measured eGFR or eGFR<sub>Cys</sub>. Using sCr and serum cystatin C (eGFR<sub>Cr-Cys</sub>) yields better concordance to measured eGFR across all races than does eGFR estimation based on Cr alone. Confirmation with CysC can avoid misdiagnosis, incorrect dosing of drugs, and inaccurate representation of the fitness for duty.</p> <p><span class="Drop">C</span>linicians usually measure renal function by using surrogate markers because directly measuring glomerular filtration rate (GFR) is not routinely feasible in a clinical setting.<sup>1,2 </sup>Creatinine (Cr) and cystatin C (CysC) are the 2 main surrogate molecules used to estimate GFR.<sup>3</sup></p> <p>Creatine is a molecule nonenzymatically converted into Cr, weighing only 113 Da in skeletal muscles.<sup>4</sup> It is then filtered at the glomeruli and secreted at the proximal tubules of the kidneys. However, serum Cr (sCr) levels are affected by several factors, including age, biological sex, liver function, diet, and muscle mass.<sup>5</sup> Historically, sCr levels also are affected by race.<sup>5</sup> In an early study of factors affecting accurate GFR, researchers reported that self-identified African American patients had a 16% higher GFR than those who did not when using Cr.<sup>6</sup> Despite this, the inclusion of Cr on a basic metabolic panel has allowed automatic reporting of an estimated GFR using sCr (eGFR<sub>Cr</sub>) to be readily available.<sup>7 <br/><br/></sup>In comparison to Cr, CysC is an endogenous protein weighing 13 kDa produced by all nucleated cells.<sup>8,9</sup> CysC is filtered by the kidney at the glomeruli and completely reabsorbed and catabolized by epithelial cells at the proximal tubule.<sup>9</sup> Since production is not dependent on skeletal muscle, there are fewer physiological impacts on serum concentration of CysC. Levels of CysC may be elevated by factors shown in the Table.</p> <h2>Estimating Glomerular Filtration Rates</h2> <p>Multiple equations were developed to mitigate the impact of extraneous factors on the accuracy of an eGFR<sub>Cr</sub>. The first widely used equation that included a variable adjustment for race was the Modification of Diet in Renal Disease study, presented in 2006.<sup>10</sup> The equation increased the accuracy of eGFR<sub>Cr</sub> further by adjusting for sex and age. It was followed by the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation in 2009, which was more accurate at higher GFR levels.<sup>11</sup> </p> <p>CysC was simultaneously studied as an alternative to Cr with multiple equation iterations shown to be viable in various populations as early as 2003.<sup>12-15</sup> However, it was not until 2012 that an equation for the use of CysC was offered for widespread use as an alternative to Cr alongside further refinement of the CKD-EPI equation for Cr.<sup>16</sup> A new formula was presented in 2021 to use both sCr and serum CysC levels to obtain a more accurate estimation of GFR.<sup>17</sup> Research continues its effort to accurately estimate GFR for diagnosing kidney disease and assessing comorbidities relating to decreased kidney function.<sup>3</sup>All historical equations attempted to mitigate the potential impact of race on sCr level when calculating eGFR<sub>Cr</sub><sup> </sup>by assigning a separate variable for African American patients. As an unintended adverse effect, these equations may have led to discrimination by having a different equation for African American patients.<sup>18 </sup>Moreover, these Cr-based equations remain less accurate in patients with varied muscle mass, such as older patients, bodybuilders, athletes, and individuals with varied extremes of daily protein intake.<sup>1,8,9,19</sup><sup> </sup>Several medications can also directly affect Cr clearance, reducing its ability to act as a surrogate for kidney function.<sup>1</sup><sup> </sup>In this case report, we discuss an African American patient with high muscle mass and protein intake who was initially diagnosed with kidney disease based on an elevated Cr and found to be misdiagnosed based on the use of CysC for a more accurate GFR estimation. </p> <h2>Case Presentation</h2> <p>A 35-year-old African American man serving in the military and recently diagnosed with HIV was referred to a nephrology clinic for further evaluation of an acute elevation in sCr. Before treatment for HIV, a brief record review showed a baseline Cr of about 1.3 mg/dL, with an eGFR<sub>Cr</sub> of 75 mL/min/1.73 m<sup>2</sup>.<sup>20</sup> In the same month, the patient was prescribed bictegravir/emtricitabine/tenofovir alafenamide, an HIV drug with nephrotoxic potential.<sup>21</sup> The patient's total viral load remained low, and CD4 count remained &gt; 500 after initiation of the HIV treatment. He was in his normal state of health and had no known contributory history before his HIV diagnosis. Cr readings peaked at 1.83 mg/dL after starting the HIV treatment<b> </b>and remained elevated to 1.73 mg/dL over the next few months, corresponding to CKD stage 3A. Because bictegravir/emtricitabine/tenofovir alafenamide is cleared by the kidneys and has a nephrotoxic profile, the clinical care team considered dosage adjustment or a medication switch given his observed elevated eGFR<sub>Cr</sub> based on the CKD-EPI 2021 equation for Cr alone. It was also noted that the patient had a similar Cr spike to 1.83<b> </b>mg/dL<b> </b>in 2018 without any identifiable renal insult or symptoms (Figure).</p> <h3>Diagnostic Evaluation </h3> <p>The primary care team ordered a renal ultrasound and referred the patient to the nephrology clinic. The nephrologist ordered the following laboratory studies: urine microalbumin to Cr ratio, basic metabolic panel (BMP), comprehensive metabolic panel (CMP), urinalysis, urine protein, urine Cr, parathyroid hormone level, hemoglobin A<sub>1c</sub>, complement component 3/4 panels, antinuclear and antineutrophil cytoplasmic antibodies titers, glomerular basement membrane antibody titer, urine light chains, serum protein electrophoresis, κ/λ ratio, viral hepatitis panel, and rapid plasma reagin testing. Much of this laboratory evaluation served to rule out any secondary causes of kidney disease, including autoimmune disease, monoclonal or polyclonal gammopathies, diabetic nephropathy or glomerulosclerosis, and nephrotic or nephritic syndromes. </p> <p>All laboratory studies returned within normal limits; no proteinuria was discovered on urinalysis, and no abnormalities were visualized on renal ultrasound. Bictegravir/emtricitabine/tenofovir alafenamide nephrotoxicity was highest among the differential diagnoses due to the timing of Cr elevation coinciding with the initiation of the medications. The patient's CysC level was 0.85 mg/dL with a calculated eGFR<sub>Cys</sub> of 125 mL/min/1.73 m<sup>2</sup>. The calculated sCR and serum cystatin C (eGFR<sub>Cr</sub><sub>-</sub><sub>Cys</sub>) using the new 2021 equation and when adjusting for body surface area placed his eGFR at 92 mL/min/1.73 m<sup>2</sup>.<sup>20</sup><sup> <br/><br/></sup>The patient’s eGFR<sub>Cys</sub><sub> </sub>reassured the care team that the patient’s renal function was not acutely or chronically impacted by bictegravir/emtricitabine/tenofovir alafenamide, resulting in avoidance of unnecessary dosage adjustment or discontinuation of the HIV treatment. The patient reported a chronic habit of protein and creatine supplementation and bodybuilding, which likely further compounded the discrepancy between eGFR<sub>Cr </sub>and eGFR<sub>Cys</sub> and explained his previous elevation in Cr in 2018.</p> <h3>Follow-up</h3> <p>The patient underwent serial monitoring that revealed a stable Cr and unremarkable eGFR, ruling out CKD. There has been no evidence of worsening kidney disease to date, and the patient remained on his initial HIV regimen.</p> <h2>Discussion</h2> <p>This case shows the importance of using CysC as an alternative or confirmatory marker compared with sCr to estimate GFR in patients with high muscle mass and/or high creatine intake, such as many in the US Department of Defense (DoD) and US Department of Veterans Affairs (VA) patient populations. In the presented case, recorded Cr levels climbed from baseline Cr with the initiation of bictegravir/emtricitabine/tenofovir alafenamide. This raised the concern that HIV treatment was leading to the development of kidney damage.<sup>22</sup> </p> <p>Diagnosis of kidney disease as opposed to the normal decline of eGFR with age in individuals without intrinsic CKD requires GFR ≥ 60 mL/min/1.73 m<sup>2</sup> with kidney damage (proteinuria or radiological abnormalities, etc) or GFR &lt; 135 to 140 mL/min/1.73 m<sup>2</sup><sup> </sup>minus the patient’s age in years.<sup>23</sup> The patient’s Cr peak at 1.83 mg/dL in 2018 led to an inappropriate diagnosis of kidney disease stage 3a based on an eGFR<sub>Cr</sub> (2021 equation) of 52 mL/min/1.73 m<sup>2</sup> when not corrected for body surface area.<sup>20</sup> However, using the new 2021 equation using both Cr and CysC, the patient’s eGFR<sub>Cr</sub><sub>-</sub><sub>Cys</sub><sub> </sub>was 92 mL/min/1.73 m<sup>2</sup> after a correction for body surface area.<br/><br/>The 2009 CKD-EPI recommended the calculation of eGFR based on SCr concentration using age, sex, and race while the 2021 CKD-EPI recommended the exclusion of race.<sup>3</sup> Both equations are less accurate in African American patients, individuals taking medications that interfere with Cr secretion and assay, and patients taking creatine supplements, high daily protein intake, or with high muscle mass.<sup>7</sup> These settings result in a decreased eGFR<sub>Cr</sub> without corresponding eGFR<sub>Cys</sub> changes. Using SCr and CysC together, the eGFR<sub>Cr-Cys </sub>yields improved concordance to measured GFR across race groups compared to GFR estimation based on Cr alone, which can avoid unnecessary expensive diagnostic workup, inappropriate kidney disease diagnosis, incorrect dosing of drugs, and accurately represent the military readiness of patients. Interestingly, in African American patients with recently diagnosed HIV, CKD-EPI using both Cr and CysC without race inclusion led to only a 2.9% overestimation of GFR and was the only equation with no statistically significant bias compared with measured GFR.<sup>24<br/><br/></sup>A March 2023 case involving an otherwise healthy 26-year-old male active-duty US Navy member with a history of excessive protein supplement intake and intense exercise &lt; 24 hours before laboratory work was diagnosed with CKD after a measured Cr of 16 mg/dL and an eGFR<sub>Cr</sub> of 4 mL/min/1.73 m<sup>2</sup> without any other evidence of kidney disease. His CysC remained within normal limits, resulting in a normal eGFR<sub>Cys</sub> of 121 mL/min/1.73 m<sup>2</sup>, indicating no CKD. His Cr and eGFR recovered 10 days after his clinic visit and cessation of his supplement intake. These findings may not be uncommon given that 65% of active-duty military use protein supplements and 38% use other performance-enhancing supplements, such as creatine, according to a study.<sup>25 <br/><br/></sup>Unfortunately, the BMP/CMP traditionally used at VA centers use the eGFR<sub>Cr </sub>equation, and it is unknown how many primary care practitioners recognize the limitations of these metabolic panels on accurate estimation of kidney function. However, in 2022 an expert panel including VA physicians recommended the immediate use of eGFR<sub>Cr-Cys </sub>or eGFR<sub>Cys</sub> for confirmatory testing and potentially screening of CKD.<sup>26</sup> A small number of VAs have since adopted this recommendation, which should lead to fewer misdiagnoses among US military members as clinicians should now have access to more accurate measurements of GFR. <br/><br/>The VA spends about $18 billion (excluding dialysis) for care for 1.1 to 2.5 million VA patients with CKD.<sup>27</sup> The majority of these diagnoses were undoubtedly made using the eGFR<sub>Cr</sub> equation, raising the question of how many may be misdiagnosed. Assessment with CysC is currently relatively expensive, but it will likely become more affordable as the use of CysC as a confirmatory test increases.<sup>5</sup> The cost of a sCr test is about $2.50, while CysC costs about $10.60, with variation from laboratory to laboratory.<sup>28</sup> By comparison, a renal ultrasound costs $99 to $140 for uninsured patients.<sup>29</sup> Furthermore, the cost of CysC testing is likely to trend downward as more facilities adopt the use of CysC measurements, which can be run on the same analytical equipment currently used for Cr measurements. Currently, most laboratories do not have established assays to use in-house and thus require CysC to be sent out to a laboratory, which increases result time and makes Cr a more attractive option. As more laboratories adopt assays for CysC, the cost of reagents will further decrease. <br/><br/>Given such considerations, confirmation testing of kidney function with CysC in specific patient populations with decreased eGFR<sub>Cr</sub> without other features of CKD can offer great medical and financial benefits. A 2023 KDIGO report noted that many individuals may be mistakenly diagnosed with CKD when using eGFR<sub>Cr</sub>.<sup>3</sup> KDIGO noted that a 2013 meta-analysis of 90,000 individuals found that with a Cr-based eGFR of 45 to 59 mL/min/1.73 m<sup>2</sup> (42%) had a CysC-based eGFR of ≥ 60 mL/min/1.73 m<sup>2</sup>. An eGFR<sub>Cr</sub> of 45 to 59 represents 54% of all patients with CKD, amounting to millions of people (including current and former military personnel).<sup>3,29-31</sup> Correcting a misdiagnosis of CKD would bring significant relief to patients and save millions in health care spending.</p> <h2>Conclusions</h2> <p>In patients who meet CKD criteria using eGFR<sub>Cr</sub> but without other features of CKD, we recommend using confirmatory CysC levels and the eGFR<sub>Cr-Cys</sub> equation. This will align care with the KDIGO guidelines and could be a cost-effective step toward improving military patient care. Further work in this area should focus on determining the knowledge gaps in primary care practitioners’ understanding of the limits of eGFR<sub>Cr</sub>, the potential mitigation of concomitant CysC testing in equivocal CKD cases, and the cost-effectiveness and increased utilization of CysC.</p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Family Medicine Residency, Eglin Air Force Base, Florida <br/><br/><sup>b</sup>Department of Internal Medicine, University of Florida, Gainesville<br/><br/><sup>c</sup>Carl T. Hayden Veterans Affairs Medical Center, Phoenix, Arizona</em> </p> <p class="isub">Author disclosures </p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article. </em> </p> <p class="isub">Disclaimer </p> <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.</em> </p> <p class="isub">Ethics and consent</p> <p> <em>Verbal and written informed consent for publication was obtained from the patient. All identifying patient information has been removed to protect patient privacy.</em> </p> <p class="isub">References</p> <p class="reference"> 1. Gabriel R. Time to scrap creatinine clearance? <i>Br Med J (Clin Res Ed).</i> 1986;293(6555):1119-1120. doi:10.1136/bmj.293.6555.1119<br/><br/> 2. Swan SK. The search continues—an ideal marker of GFR. <i>Clin Chem</i>. 1997;43(6):913-914.doi:10.1093/clinchem/43.6.913 3. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. <i>Kidney Int Suppl</i>. 2013;3(1).<br/><br/> 4. Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabolism. <i>Physiol Rev</i>. 2000;80(3):1107-1213. doi:10.1152/physrev.2000.80.3.1107<br/><br/> 5. Ferguson TW, Komenda P, Tangri N. Cystatin C as a biomarker for estimating glomerular filtration rate. <i>Curr Opin Nephrol Hypertens</i>. 2015;24(3):295-300. doi:10.1097/mnh.0000000000000115<br/><br/> 6. Levey AS, Titan SM, Powe NR, Coresh J, Inker LA. Kidney disease, race, and GFR estimation. <i>Clin J Am Soc Nephrol</i>. 2020;15(8):1203-1212. doi:10.2215/cjn.12791019<br/><br/> 7. Shlipak MG, Tummalapalli SL, Boulware LE, et al; Conference Participants. The case for early identification and intervention of chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) controversies conference. <i>Kidney Int</i>. 2021;99(1):34-47. doi:10.1016/j.kint.2020.10.012<br/><br/> 8. O’Riordan SE, Webb MC, Stowe HJ, et al. Cystatin C improves the detection of mild renal dysfunction in older patients. <i>Ann Clin Biochem</i>. 2003;40(pt 6):648-655. doi:10.1258/000456303770367243<br/><br/> 9. Stevens LA, Schmid CH, Greene T, et al. Factors other than glomerular filtration rate affect serum cystatin C levels. <i>Kidney Int</i>. 2009;75(6):652-660. doi:10.1038/ki.2008.638<br/><br/>10. Levey AS, Coresh J, Greene T, et al; Chronic Kidney Disease Epidemiology Collaboration. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. <i>Ann Intern Med</i>. 2006;145(4):247-254. doi:10.7326/0003-4819-145-4-200608150-00004<br/><br/>11. Levey AS, Stevens LA, Schmid CH, et al; Chronic Kidney Disease Epidemiology Collaboration. A new equation to estimate glomerular filtration rate. <i>Ann Intern Med</i>. 2009;150(9):604-612. doi:10.7326/0003-4819-150-9-200905050-00006<br/><br/>12. Pöge U, Gerhardt T, Stoffel-Wagner B, Klehr HU, Sauerbruch T, Woitas RP. Calculation of glomerular filtration rate based on cystatin C in cirrhotic patients. <i>Nephrol Dial Transplant</i>. 2006;21(3):660-664. doi:10.1093/ndt/gfi305<br/><br/>13. Larsson A, Malm J, Grubb A, Hansson LO. Calculation of glomerular filtration rate expressed in mL/min from plasma cystatin C values in mg/L. <i>Scand J Clin Lab Invest</i>. 2004;64(1):25-30. doi:10.1080/00365510410003723. <br/><br/>14. Macisaac RJ, Tsalamandris C, Thomas MC, et al. Estimating glomerular filtration rate in diabetes: a comparison of cystatin-C- and creatinine-based methods. <i>Diabetologia</i>. 2006;49(7):1686-1689. doi:10.1007/s00125-006-0275-7<br/><br/>15. Rule AD, Bergstralh EJ, Slezak JM, Bergert J, Larson TS. Glomerular filtration rate estimated by cystatin C among different clinical presentations. <i>Kidney Int</i>. 2006;69(2):399-405. doi:10.1038/sj.ki.5000073<br/><br/>16. Inker LA, Schmid CH, Tighiouart H, et al; Chronic Kidney Disease Epidemiology Collaboration Investigators. Estimating glomerular filtration rate from serum creatinine and cystatin C. <i>N Engl J Med</i>. 2012;367(1):20-29. doi:10.1056/NEJMoa1114248<br/><br/>17. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. <i>N Engl J Med</i>. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234<br/><br/>18. Inker LA, Eneanya ND, Coresh J, et al; Chronic Kidney Disease Epidemiology Collaboration. New creatinine- and cystatin C–Based equations to estimate GFR without race. <i>N Engl J Med</i>. 2021;385(19):1737-1749. doi:10.1056/NEJMoa2102953<br/><br/>19. Oterdoom LH, Gansevoort RT, Schouten JP, de Jong PE, Gans ROB, Bakker SJL. Urinary creatinine excretion, an indirect measure of muscle mass, is an independent predictor of cardiovascular disease and mortality in the general population. <i>Atherosclerosis</i>. 2009;207(2):534-540. doi.10.1016/j.atherosclerosis.2009.05.010<br/><br/>20. National Kidney Foundation Inc. eGFR calculator. Accessed October 20, 2023. https://www.kidney.org/professionals/kdoqi/gfr_calculator<br/><br/>21. Ueaphongsukkit T, Gatechompol S, Avihingsanon A, et al. Tenofovir alafenamide nephrotoxicity: a case report and literature review. <i>AIDS Res Ther</i>. 2021;18(1):53. doi:10.1186/s12981-021-00380-w<br/><br/>22. D’Agati V, Appel GB. Renal pathology of human immunodeficiency virus infection. <i>Semin Nephrol</i>. 1998;18(4):406-421.<br/><br/>23. Glassock RJ, Winearls C. Ageing and the glomerular filtration rate: truths and consequences. <i>Trans Am Clin Climatol Assoc</i>. 2009;120:419-428. <br/><br/>24. Seape T, Gounden V, van Deventer HE, Candy GP, George JA. Cystatin C- and creatinine-based equations in the assessment of renal function in HIV-positive patients prior to commencing highly active antiretroviral therapy<i>. Ann Clin Biochem</i>. 2016;53(pt 1):58-66. doi:10.1177/0004563215579695<br/><br/>25. Tobin TW, Thurlow JS, Yuan CM. A healthy active duty soldier with an elevated serum creatinine. <i>Mil Med</i>. 2023;188(3-4):e866-e869. doi:10.1093/milmed/usab163<br/><br/>26. Delgado C, Baweja M, Crews DC, et al. A unifying approach for GFR estimation: recommendations of the NKF-ASN Task Force on Reassessing the Inclusion of Race in Diagnosing Kidney Disease. <i>Am J Kidney Dis</i>. 2022;79(2):268-288.e1. doi:10.1053/j.ajkd.2021.08.003<br/><br/>27. Saran R, Pearson A, Tilea A, et al; VA-REINS Steering Committee; VA Advisory Board. Burden and cost of caring for us veterans with CKD: initial findings from the VA Renal Information System (VA-REINS). <i>Am J Kidney Dis</i>. 2021;77(3):397-405. doi:10.1053/j.ajkd.2020.07.013<br/><br/>28. Zoler ML. Nephrologists make the case for cystatin C-based eGFR. Accessed October 20, 2023. https://www.medscape.com/viewarticle/951335#vp_2 <br/><br/>29. Versaw N. How much does an ultrasound cost? Updated February 2022. Accessed October 20, 2023. https://www.compare.com/health/healthcare-resources/how-much-does-an-ultrasound-cost<br/><br/>30. Levey AS, Coresh J. Chronic kidney disease. <i>Lancet</i>. 2012;379(9811):165-180. doi:10.1016/S0140-6736(11)60178-5<br/><br/>31. Shlipak MG, Matsushita K, Ärnlöv J, et al; CKD Prognosis Consortium. Cystatin C versus creatinine in determining risk based on kidney function. <i>N Engl J Med</i>. 2013;369(10):932-943. doi:10.1056/NEJMoa1214234</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

Thiazide-Induced Hyponatremia Presenting as a Fall in an Older Adult

Article Type
Changed
Sat, 02/03/2024 - 22:33

Hypertension is a major risk factor for heart disease, stroke, and kidney disease.The prevalence of hypertension increases with age, primarily due to age-related changes in arterial physiology.1 For older adults, current guidelines regarding blood pressure (BP) treatment goals vary. The American Heart Association/American College of Cardiology 2017 clinical practice guidelines recommend a systolic BP (SBP) treatment goal of < 130 mm Hg for community-dwelling, ambulatory, noninstitutionalized adults aged ≥ 65 years; whereas the American College of Physicians/American Academy of Family Physicians recommend a goal of < 150 mm Hg for those aged ≥ 60 years without comorbidities and < 140 mm Hg for those with increased cardiovascular risk.1-3 Regardless of the specific threshold, agreement that some degree of BP control even in those with advanced age improves outcomes.2

First-line therapy for uncomplicated hypertension includes thiazide diuretics, long-acting calcium channel blockers, and renin-angiotensin system inhibitors. When choosing between these options, it is recommended to engage in shared decision making and to consider the patient’s comorbidities. Among patients who are likely to require a second agent (eg, if initial BP is > 20/10 mm Hg above goal), it is recommended to begin both drugs at the same time, preferably benazepril plus amlodipine due to the reduction in cardiovascular events reported in the ACCOMPLISH trial.4 If BP remains elevated despite 2 agents at moderate to maximum doses, it is important to investigate for secondary hypertension causes and to explore medication adherence as possible etiologies of treatment failure. Older adults are often at higher risk of adverse drug events due to age-related changes in pharmacodynamics. Despite this, there are no guidelines for choosing between different classes of antihypertensives in this population. We present a case of thiazide-induced hyponatremia in an older adult and review the risks of thiazide use in this population.

Case Presentation

A man aged > 90 years was admitted to the hospital after a syncopal episode. His history was significant for hypertension, hyperlipidemia, and vitamin D deficiency. At the time, his home medications were amlodipine 5 mg daily, atorvastatin 40 mg daily, ergocalciferol 50,000 IU weekly, and polyethylene glycol 17 g daily as needed. His syncope workup was unremarkable and included negative orthostatic vital signs, normal serial troponins, an electrocardiogram without ischemic changes, normal serum creatinine, sodium, and glucose, and a head computed tomography without any acute abnormality. Throughout the patient’s hospital stay, he had multiple elevated SBP readings, including many > 200 mm Hg. On discharge, in addition to continuing his home medications, he was started on valsartan 20 mg daily and enrolled in a remote BP monitoring program.

Three weeks later, the patient was seen by their primary care practitioner for follow-up. He reported adherence to his antihypertensive regimen. However, his remote BP monitoring revealed persistently elevated BPs, with an average of 179/79 mm Hg, a high of 205/85 mm Hg, and a low of 150/67 mm Hg over the previous 7 days. Laboratory tests obtained at the visit were notable for serum sodium of 138 mmol/L and potassium of 4.1 mmol/L. His weight was 87 kg. Given persistently elevated BP readings, in addition to continuing his amlodipine 5 mg daily and valsartan 20 mg daily, he was started on hydrochlorothiazide 25 mg daily, with plans to repeat a basic metabolic panel in 2 weeks.

Two weeks later, he fell after getting out of his bed. On examination, he was noted to have dry mucous membranes, and although no formal delirium screening was performed, he was able to repeat the months of the year backward. Vital signs were notable for positive postural hypertension, and his laboratory tests revealed a normal serum creatinine, serum sodium of 117 mmol/L (reference range, 135-145 mmol/L), serum potassium of 3.2 mmol/L (reference range, 3.0-5.0 mmol/L), a low serum osmolality, and urine sodium of 35 mmol/L most consistent with hypovolemic hypoosmotic hyponatremia secondary to thiazide initiation. The patient’s hydrochlorothiazide was discontinued, and he was admitted to the hospital for close monitoring. His sodium levels gradually normalized over the next 2 weeks without any other intervention.

Discussion

Although thiazide diuretics are recommended as first-line therapy for uncomplicated hypertension, they are known to cause electrolyte abnormalities, including hypomagnesemia, hypokalemia, and hyponatremia.4 These metabolic derangements are more likely to occur in older adults. One study of adults aged ≥ 65 years found that at 9 months of follow-up, 14.3% of new thiazide users had developed a thiazide-related metabolic adverse event (hyponatremia < 135 mmol/L, hypokalemia < 3.5 mmol/L, and decrease in estimated glomerular filtration rate by > 25%) compared with 6.0% of nonusers (P < .001; number needed to harm [NNH] = 12).5 In addition, 3.8% of new thiazide users had an emergency department visit or were hospitalized for complications related to thiazides compared with only 2.0% of nonusers (P = .02; NNH = 56).5 Independent risk factors for thiazide-induced hyponatremia include high-comorbidity burden, low body weight, low-normal or unmeasured serum sodium, low potassium, and aged > 70 years.5-7 Each 10-year increment in age is associated with a 2-fold increase in risk, suggesting that older adults are at a much higher risk for hyponatremia than their younger peers.6

Despite their designation as a first-line option for uncomplicated hypertension, thiazide diuretics may cause more harm than good in some older adults, especially those with additional risk factors for thiazide-induced hyponatremia. In this population, these adverse effects should be discussed before starting thiazides for the treatment of hypertension. If thiazides are initiated, they should be started at the lowest possible dose, and plans made to monitor bloodwork within 1 to 2 weeks of initiation or dose change and periodically thereafter while the patient remains on the therapy.

 

 

Medication Management in Older Adults

Due to the risks of medication use in older adults, the phrase “start low, go slow” is commonly used in geriatric medicine to describe the optimal method for initiation and up-titration of new medication with the hope of mitigating adverse drug events. In our case, we started valsartan at 20 mg daily—one-fourth the recommended initial dose. Although this strategy is reasonable to “start low,” we were not surprised to find that the patient’s BP did not markedly improve on such a low dose. The team could have increased the valsartan dose to a therapeutically efficacious dose before choosing to add another hypertensive agent. In alignment with geriatric prescribing principles, starting at the lowest possible dose of hydrochlorothiazide is recommended.5 However, the clinician started hydrochlorothiazide at 25 mg daily, potentially increasing this patient’s risk of electrolyte abnormalities and eventual fall.

Managing hypertension also invites a discussion of polypharmacy and medication adherence. Older adults are at risk of polypharmacy, defined as the prescription of 5 or more medications.8 Polypharmacy is associated with increased hospitalizations, higher costs of care for individuals and health care systems, increased risks of adverse drug events, medication nonadherence, and lower quality of life for patients.9 In some situations, the risks of polypharmacy may outweigh the benefits of using multiple antihypertensives with different mechanisms of action if patients can reach their BP goal on the maximum dose of a single agent. For patients taking multiple antihypertensives, it is important to routinely monitor BP and assess whether deprescribing is indicated. Cognitive impairment and decreased social support may affect medication adherence for older adults.6 Clinicians should be aware of strategies, such as medication reminders and pillboxes, to increase antihypertensive medication adherence. Polypills that contain 2 antihypertensives can be another tool used to manage older adults to increase adherence and decrease health care costs.10

figure.png

A current strategy that encompasses discussing many, if not all, of these noted elements is the Institute for Healthcare Improvement’s Age-Friendly Health System. This framework uses evidence-based tools to provide care for older adults across all clinical settings and highlights the 4Ms: what matters, medication, mentation, and mobility.11 Medication considers whether a medication is necessary, whether its use has benefits that outweigh the risks, and how it interacts with what matters, mentation, and mobility. In particular, what matters plays an important role in hypertension management in older adults given the recommended target BP differs, depending on which specialty organization guideline is followed. By better understanding what matters to patients, including their goals and priorities, clinicians can engage patients in shared decision making and provide individualized recommendations based on geriatric principles (eg, start low, go slow, principles of medication adherence) and patient comorbidities (eg, medical history and risk factors for hyponatremia) to help patients make a more informed choice about their antihypertensive treatment regimen (Figure).

Conclusions

This case illustrates the need for a specialized approach to hypertension management in older adults and the risks of thiazide diuretics in this population. Clinicians should consider BP goals, patient-specific factors, and principles of medication management in older adults. If initiating thiazide therapy, discuss the risks associated with use, start at the lowest possible dose, and monitor bloodwork within 1 to 2 weeks of initiation/dose change and periodically thereafter while the patient remains on the therapy to decrease the risk of adverse events. Finally, the Institute for Healthcare Improvement’s Age-Friendly Health System framework can be a useful when considering the addition of a new medication in an older adult’s treatment plan.

Acknowledgments

This material is the result of work supported with resources and the use of facilities at the New England Geriatrics Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, and the Cincinnati VeteransAffairs Medical Center.

References

1. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2018;71(19):e127-e248. doi:10.1016/j.jacc.2017.11.006

2. Davis LL. Hypertension: how low to go when treating older adults. J Nurse Pract. 2019;15(1):1-6. doi:10.1016/j.nurpra.2018.10.010

3. Qaseem A, Wilt TJ, Rich R, et al. Pharmacologic Treatment of Hypertension in Adults Aged 60 Years or Older to Higher Versus Lower Blood Pressure Targets: A Clinical Practice Guideline From the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med. 2017;166(6):430-437. doi:10.7326/M16-1785

4. Aronow WS, Fleg JL, Pepine CJ, et al. ACCF/AHA 2011 expert consensus document on hypertension in the elderly: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus documents developed in collaboration with the American Academy of Neurology, American Geriatrics Society, American Society for Preventive Cardiology, American Society of Hypertension, American Society of Nephrology, Association of Black Cardiologists, and European Society of Hypertension. J Am Coll Cardiol. 2011;57(20):2037-2114. doi:10.1016/j.jacc.2011.01.008

5. Makam AN, Boscardin WJ, Miao Y, Steinman MA. Risk of thiazide-induced metabolic adverse events in older adults. J Am Geriatr Soc. 2014;62(6):1039-1045. doi:10.1111/jgs.12839

6. Chow KM, Szeto CC, Wong TY, Leung CB, Li PK. Risk factors for thiazide-induced hyponatraemia. QJM. 2003;96(12):911-917. doi:10.1093/qjmed/hcg157

7. Clayton JA, Rodgers S, Blakey J, Avery A, Hall IP. Thiazide diuretic prescription and electrolyte abnormalities in primary care. Br J Clin Pharmacol. 2006;61(1):87-95. doi:10.1111/j.1365-2125.2005.02531.x

8. Shah BM, Hajjar ER. Polypharmacy, adverse drug reactions, and geriatric syndromes. Clin Geriatr Med. 2012;28(2):173-186. doi:10.1016/j.cger.2012.01.002

9. Benetos A, Petrovic M, Strandberg T. Hypertension management in older and frail older patients. Circ Res. 2019;124(7):1045-1060. doi:10.1161/CIRCRESAHA.118.313236

10. Sherrill B, Halpern M, Khan S, Zhang J, Panjabi S. Single-pill vs free-equivalent combination therapies for hypertension: a meta-analysis of health care costs and adherence. J Clin Hypertens (Greenwich). 2011;13(12):898-909. doi:10.1111/j.1751-7176.2011.00550.x

11. Mate K, Fulmer T, Pelton L, et al. Evidence for the 4Ms: interactions and outcomes across the care continuum. J Aging Health. 2021;33(7-8):469-481. doi:10.1177/0898264321991658

Article PDF
Author and Disclosure Information

Stephanie Pagliuca, MDa,b; Caroline Wagner, PharmDc; Brittany L. Pietruszka, PharmDc; Shivani K. Jindal, MD, MPHb,c,d

Correspondence:  Shivani K. Jindal  (shivani.jindal@va.gov)

aNew England Geriatrics Research, Education, and Clinical Center (GRECC), Veterans Affairs Boston Healthcare System, Massachusetts

bBoston University Chobanian & Avedisian School of Medicine, Massachusetts

cVeterans Affairs Boston Healthcare System, Massachusetts

dCincinnati Veterans Affairs Medical Center, Ohio

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 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

This manuscript has been reviewed by the Veterans Affairs Boston Privacy Office prior to submission. We have received written consent and release of information from this veteran to use details of their case for this manuscript.

Issue
Federal Practitioner - 41(2)a
Publications
Topics
Page Number
58
Sections
Author and Disclosure Information

Stephanie Pagliuca, MDa,b; Caroline Wagner, PharmDc; Brittany L. Pietruszka, PharmDc; Shivani K. Jindal, MD, MPHb,c,d

Correspondence:  Shivani K. Jindal  (shivani.jindal@va.gov)

aNew England Geriatrics Research, Education, and Clinical Center (GRECC), Veterans Affairs Boston Healthcare System, Massachusetts

bBoston University Chobanian & Avedisian School of Medicine, Massachusetts

cVeterans Affairs Boston Healthcare System, Massachusetts

dCincinnati Veterans Affairs Medical Center, Ohio

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 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

This manuscript has been reviewed by the Veterans Affairs Boston Privacy Office prior to submission. We have received written consent and release of information from this veteran to use details of their case for this manuscript.

Author and Disclosure Information

Stephanie Pagliuca, MDa,b; Caroline Wagner, PharmDc; Brittany L. Pietruszka, PharmDc; Shivani K. Jindal, MD, MPHb,c,d

Correspondence:  Shivani K. Jindal  (shivani.jindal@va.gov)

aNew England Geriatrics Research, Education, and Clinical Center (GRECC), Veterans Affairs Boston Healthcare System, Massachusetts

bBoston University Chobanian & Avedisian School of Medicine, Massachusetts

cVeterans Affairs Boston Healthcare System, Massachusetts

dCincinnati Veterans Affairs Medical Center, Ohio

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 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

This manuscript has been reviewed by the Veterans Affairs Boston Privacy Office prior to submission. We have received written consent and release of information from this veteran to use details of their case for this manuscript.

Article PDF
Article PDF

Hypertension is a major risk factor for heart disease, stroke, and kidney disease.The prevalence of hypertension increases with age, primarily due to age-related changes in arterial physiology.1 For older adults, current guidelines regarding blood pressure (BP) treatment goals vary. The American Heart Association/American College of Cardiology 2017 clinical practice guidelines recommend a systolic BP (SBP) treatment goal of < 130 mm Hg for community-dwelling, ambulatory, noninstitutionalized adults aged ≥ 65 years; whereas the American College of Physicians/American Academy of Family Physicians recommend a goal of < 150 mm Hg for those aged ≥ 60 years without comorbidities and < 140 mm Hg for those with increased cardiovascular risk.1-3 Regardless of the specific threshold, agreement that some degree of BP control even in those with advanced age improves outcomes.2

First-line therapy for uncomplicated hypertension includes thiazide diuretics, long-acting calcium channel blockers, and renin-angiotensin system inhibitors. When choosing between these options, it is recommended to engage in shared decision making and to consider the patient’s comorbidities. Among patients who are likely to require a second agent (eg, if initial BP is > 20/10 mm Hg above goal), it is recommended to begin both drugs at the same time, preferably benazepril plus amlodipine due to the reduction in cardiovascular events reported in the ACCOMPLISH trial.4 If BP remains elevated despite 2 agents at moderate to maximum doses, it is important to investigate for secondary hypertension causes and to explore medication adherence as possible etiologies of treatment failure. Older adults are often at higher risk of adverse drug events due to age-related changes in pharmacodynamics. Despite this, there are no guidelines for choosing between different classes of antihypertensives in this population. We present a case of thiazide-induced hyponatremia in an older adult and review the risks of thiazide use in this population.

Case Presentation

A man aged > 90 years was admitted to the hospital after a syncopal episode. His history was significant for hypertension, hyperlipidemia, and vitamin D deficiency. At the time, his home medications were amlodipine 5 mg daily, atorvastatin 40 mg daily, ergocalciferol 50,000 IU weekly, and polyethylene glycol 17 g daily as needed. His syncope workup was unremarkable and included negative orthostatic vital signs, normal serial troponins, an electrocardiogram without ischemic changes, normal serum creatinine, sodium, and glucose, and a head computed tomography without any acute abnormality. Throughout the patient’s hospital stay, he had multiple elevated SBP readings, including many > 200 mm Hg. On discharge, in addition to continuing his home medications, he was started on valsartan 20 mg daily and enrolled in a remote BP monitoring program.

Three weeks later, the patient was seen by their primary care practitioner for follow-up. He reported adherence to his antihypertensive regimen. However, his remote BP monitoring revealed persistently elevated BPs, with an average of 179/79 mm Hg, a high of 205/85 mm Hg, and a low of 150/67 mm Hg over the previous 7 days. Laboratory tests obtained at the visit were notable for serum sodium of 138 mmol/L and potassium of 4.1 mmol/L. His weight was 87 kg. Given persistently elevated BP readings, in addition to continuing his amlodipine 5 mg daily and valsartan 20 mg daily, he was started on hydrochlorothiazide 25 mg daily, with plans to repeat a basic metabolic panel in 2 weeks.

Two weeks later, he fell after getting out of his bed. On examination, he was noted to have dry mucous membranes, and although no formal delirium screening was performed, he was able to repeat the months of the year backward. Vital signs were notable for positive postural hypertension, and his laboratory tests revealed a normal serum creatinine, serum sodium of 117 mmol/L (reference range, 135-145 mmol/L), serum potassium of 3.2 mmol/L (reference range, 3.0-5.0 mmol/L), a low serum osmolality, and urine sodium of 35 mmol/L most consistent with hypovolemic hypoosmotic hyponatremia secondary to thiazide initiation. The patient’s hydrochlorothiazide was discontinued, and he was admitted to the hospital for close monitoring. His sodium levels gradually normalized over the next 2 weeks without any other intervention.

Discussion

Although thiazide diuretics are recommended as first-line therapy for uncomplicated hypertension, they are known to cause electrolyte abnormalities, including hypomagnesemia, hypokalemia, and hyponatremia.4 These metabolic derangements are more likely to occur in older adults. One study of adults aged ≥ 65 years found that at 9 months of follow-up, 14.3% of new thiazide users had developed a thiazide-related metabolic adverse event (hyponatremia < 135 mmol/L, hypokalemia < 3.5 mmol/L, and decrease in estimated glomerular filtration rate by > 25%) compared with 6.0% of nonusers (P < .001; number needed to harm [NNH] = 12).5 In addition, 3.8% of new thiazide users had an emergency department visit or were hospitalized for complications related to thiazides compared with only 2.0% of nonusers (P = .02; NNH = 56).5 Independent risk factors for thiazide-induced hyponatremia include high-comorbidity burden, low body weight, low-normal or unmeasured serum sodium, low potassium, and aged > 70 years.5-7 Each 10-year increment in age is associated with a 2-fold increase in risk, suggesting that older adults are at a much higher risk for hyponatremia than their younger peers.6

Despite their designation as a first-line option for uncomplicated hypertension, thiazide diuretics may cause more harm than good in some older adults, especially those with additional risk factors for thiazide-induced hyponatremia. In this population, these adverse effects should be discussed before starting thiazides for the treatment of hypertension. If thiazides are initiated, they should be started at the lowest possible dose, and plans made to monitor bloodwork within 1 to 2 weeks of initiation or dose change and periodically thereafter while the patient remains on the therapy.

 

 

Medication Management in Older Adults

Due to the risks of medication use in older adults, the phrase “start low, go slow” is commonly used in geriatric medicine to describe the optimal method for initiation and up-titration of new medication with the hope of mitigating adverse drug events. In our case, we started valsartan at 20 mg daily—one-fourth the recommended initial dose. Although this strategy is reasonable to “start low,” we were not surprised to find that the patient’s BP did not markedly improve on such a low dose. The team could have increased the valsartan dose to a therapeutically efficacious dose before choosing to add another hypertensive agent. In alignment with geriatric prescribing principles, starting at the lowest possible dose of hydrochlorothiazide is recommended.5 However, the clinician started hydrochlorothiazide at 25 mg daily, potentially increasing this patient’s risk of electrolyte abnormalities and eventual fall.

Managing hypertension also invites a discussion of polypharmacy and medication adherence. Older adults are at risk of polypharmacy, defined as the prescription of 5 or more medications.8 Polypharmacy is associated with increased hospitalizations, higher costs of care for individuals and health care systems, increased risks of adverse drug events, medication nonadherence, and lower quality of life for patients.9 In some situations, the risks of polypharmacy may outweigh the benefits of using multiple antihypertensives with different mechanisms of action if patients can reach their BP goal on the maximum dose of a single agent. For patients taking multiple antihypertensives, it is important to routinely monitor BP and assess whether deprescribing is indicated. Cognitive impairment and decreased social support may affect medication adherence for older adults.6 Clinicians should be aware of strategies, such as medication reminders and pillboxes, to increase antihypertensive medication adherence. Polypills that contain 2 antihypertensives can be another tool used to manage older adults to increase adherence and decrease health care costs.10

figure.png

A current strategy that encompasses discussing many, if not all, of these noted elements is the Institute for Healthcare Improvement’s Age-Friendly Health System. This framework uses evidence-based tools to provide care for older adults across all clinical settings and highlights the 4Ms: what matters, medication, mentation, and mobility.11 Medication considers whether a medication is necessary, whether its use has benefits that outweigh the risks, and how it interacts with what matters, mentation, and mobility. In particular, what matters plays an important role in hypertension management in older adults given the recommended target BP differs, depending on which specialty organization guideline is followed. By better understanding what matters to patients, including their goals and priorities, clinicians can engage patients in shared decision making and provide individualized recommendations based on geriatric principles (eg, start low, go slow, principles of medication adherence) and patient comorbidities (eg, medical history and risk factors for hyponatremia) to help patients make a more informed choice about their antihypertensive treatment regimen (Figure).

Conclusions

This case illustrates the need for a specialized approach to hypertension management in older adults and the risks of thiazide diuretics in this population. Clinicians should consider BP goals, patient-specific factors, and principles of medication management in older adults. If initiating thiazide therapy, discuss the risks associated with use, start at the lowest possible dose, and monitor bloodwork within 1 to 2 weeks of initiation/dose change and periodically thereafter while the patient remains on the therapy to decrease the risk of adverse events. Finally, the Institute for Healthcare Improvement’s Age-Friendly Health System framework can be a useful when considering the addition of a new medication in an older adult’s treatment plan.

Acknowledgments

This material is the result of work supported with resources and the use of facilities at the New England Geriatrics Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, and the Cincinnati VeteransAffairs Medical Center.

Hypertension is a major risk factor for heart disease, stroke, and kidney disease.The prevalence of hypertension increases with age, primarily due to age-related changes in arterial physiology.1 For older adults, current guidelines regarding blood pressure (BP) treatment goals vary. The American Heart Association/American College of Cardiology 2017 clinical practice guidelines recommend a systolic BP (SBP) treatment goal of < 130 mm Hg for community-dwelling, ambulatory, noninstitutionalized adults aged ≥ 65 years; whereas the American College of Physicians/American Academy of Family Physicians recommend a goal of < 150 mm Hg for those aged ≥ 60 years without comorbidities and < 140 mm Hg for those with increased cardiovascular risk.1-3 Regardless of the specific threshold, agreement that some degree of BP control even in those with advanced age improves outcomes.2

First-line therapy for uncomplicated hypertension includes thiazide diuretics, long-acting calcium channel blockers, and renin-angiotensin system inhibitors. When choosing between these options, it is recommended to engage in shared decision making and to consider the patient’s comorbidities. Among patients who are likely to require a second agent (eg, if initial BP is > 20/10 mm Hg above goal), it is recommended to begin both drugs at the same time, preferably benazepril plus amlodipine due to the reduction in cardiovascular events reported in the ACCOMPLISH trial.4 If BP remains elevated despite 2 agents at moderate to maximum doses, it is important to investigate for secondary hypertension causes and to explore medication adherence as possible etiologies of treatment failure. Older adults are often at higher risk of adverse drug events due to age-related changes in pharmacodynamics. Despite this, there are no guidelines for choosing between different classes of antihypertensives in this population. We present a case of thiazide-induced hyponatremia in an older adult and review the risks of thiazide use in this population.

Case Presentation

A man aged > 90 years was admitted to the hospital after a syncopal episode. His history was significant for hypertension, hyperlipidemia, and vitamin D deficiency. At the time, his home medications were amlodipine 5 mg daily, atorvastatin 40 mg daily, ergocalciferol 50,000 IU weekly, and polyethylene glycol 17 g daily as needed. His syncope workup was unremarkable and included negative orthostatic vital signs, normal serial troponins, an electrocardiogram without ischemic changes, normal serum creatinine, sodium, and glucose, and a head computed tomography without any acute abnormality. Throughout the patient’s hospital stay, he had multiple elevated SBP readings, including many > 200 mm Hg. On discharge, in addition to continuing his home medications, he was started on valsartan 20 mg daily and enrolled in a remote BP monitoring program.

Three weeks later, the patient was seen by their primary care practitioner for follow-up. He reported adherence to his antihypertensive regimen. However, his remote BP monitoring revealed persistently elevated BPs, with an average of 179/79 mm Hg, a high of 205/85 mm Hg, and a low of 150/67 mm Hg over the previous 7 days. Laboratory tests obtained at the visit were notable for serum sodium of 138 mmol/L and potassium of 4.1 mmol/L. His weight was 87 kg. Given persistently elevated BP readings, in addition to continuing his amlodipine 5 mg daily and valsartan 20 mg daily, he was started on hydrochlorothiazide 25 mg daily, with plans to repeat a basic metabolic panel in 2 weeks.

Two weeks later, he fell after getting out of his bed. On examination, he was noted to have dry mucous membranes, and although no formal delirium screening was performed, he was able to repeat the months of the year backward. Vital signs were notable for positive postural hypertension, and his laboratory tests revealed a normal serum creatinine, serum sodium of 117 mmol/L (reference range, 135-145 mmol/L), serum potassium of 3.2 mmol/L (reference range, 3.0-5.0 mmol/L), a low serum osmolality, and urine sodium of 35 mmol/L most consistent with hypovolemic hypoosmotic hyponatremia secondary to thiazide initiation. The patient’s hydrochlorothiazide was discontinued, and he was admitted to the hospital for close monitoring. His sodium levels gradually normalized over the next 2 weeks without any other intervention.

Discussion

Although thiazide diuretics are recommended as first-line therapy for uncomplicated hypertension, they are known to cause electrolyte abnormalities, including hypomagnesemia, hypokalemia, and hyponatremia.4 These metabolic derangements are more likely to occur in older adults. One study of adults aged ≥ 65 years found that at 9 months of follow-up, 14.3% of new thiazide users had developed a thiazide-related metabolic adverse event (hyponatremia < 135 mmol/L, hypokalemia < 3.5 mmol/L, and decrease in estimated glomerular filtration rate by > 25%) compared with 6.0% of nonusers (P < .001; number needed to harm [NNH] = 12).5 In addition, 3.8% of new thiazide users had an emergency department visit or were hospitalized for complications related to thiazides compared with only 2.0% of nonusers (P = .02; NNH = 56).5 Independent risk factors for thiazide-induced hyponatremia include high-comorbidity burden, low body weight, low-normal or unmeasured serum sodium, low potassium, and aged > 70 years.5-7 Each 10-year increment in age is associated with a 2-fold increase in risk, suggesting that older adults are at a much higher risk for hyponatremia than their younger peers.6

Despite their designation as a first-line option for uncomplicated hypertension, thiazide diuretics may cause more harm than good in some older adults, especially those with additional risk factors for thiazide-induced hyponatremia. In this population, these adverse effects should be discussed before starting thiazides for the treatment of hypertension. If thiazides are initiated, they should be started at the lowest possible dose, and plans made to monitor bloodwork within 1 to 2 weeks of initiation or dose change and periodically thereafter while the patient remains on the therapy.

 

 

Medication Management in Older Adults

Due to the risks of medication use in older adults, the phrase “start low, go slow” is commonly used in geriatric medicine to describe the optimal method for initiation and up-titration of new medication with the hope of mitigating adverse drug events. In our case, we started valsartan at 20 mg daily—one-fourth the recommended initial dose. Although this strategy is reasonable to “start low,” we were not surprised to find that the patient’s BP did not markedly improve on such a low dose. The team could have increased the valsartan dose to a therapeutically efficacious dose before choosing to add another hypertensive agent. In alignment with geriatric prescribing principles, starting at the lowest possible dose of hydrochlorothiazide is recommended.5 However, the clinician started hydrochlorothiazide at 25 mg daily, potentially increasing this patient’s risk of electrolyte abnormalities and eventual fall.

Managing hypertension also invites a discussion of polypharmacy and medication adherence. Older adults are at risk of polypharmacy, defined as the prescription of 5 or more medications.8 Polypharmacy is associated with increased hospitalizations, higher costs of care for individuals and health care systems, increased risks of adverse drug events, medication nonadherence, and lower quality of life for patients.9 In some situations, the risks of polypharmacy may outweigh the benefits of using multiple antihypertensives with different mechanisms of action if patients can reach their BP goal on the maximum dose of a single agent. For patients taking multiple antihypertensives, it is important to routinely monitor BP and assess whether deprescribing is indicated. Cognitive impairment and decreased social support may affect medication adherence for older adults.6 Clinicians should be aware of strategies, such as medication reminders and pillboxes, to increase antihypertensive medication adherence. Polypills that contain 2 antihypertensives can be another tool used to manage older adults to increase adherence and decrease health care costs.10

figure.png

A current strategy that encompasses discussing many, if not all, of these noted elements is the Institute for Healthcare Improvement’s Age-Friendly Health System. This framework uses evidence-based tools to provide care for older adults across all clinical settings and highlights the 4Ms: what matters, medication, mentation, and mobility.11 Medication considers whether a medication is necessary, whether its use has benefits that outweigh the risks, and how it interacts with what matters, mentation, and mobility. In particular, what matters plays an important role in hypertension management in older adults given the recommended target BP differs, depending on which specialty organization guideline is followed. By better understanding what matters to patients, including their goals and priorities, clinicians can engage patients in shared decision making and provide individualized recommendations based on geriatric principles (eg, start low, go slow, principles of medication adherence) and patient comorbidities (eg, medical history and risk factors for hyponatremia) to help patients make a more informed choice about their antihypertensive treatment regimen (Figure).

Conclusions

This case illustrates the need for a specialized approach to hypertension management in older adults and the risks of thiazide diuretics in this population. Clinicians should consider BP goals, patient-specific factors, and principles of medication management in older adults. If initiating thiazide therapy, discuss the risks associated with use, start at the lowest possible dose, and monitor bloodwork within 1 to 2 weeks of initiation/dose change and periodically thereafter while the patient remains on the therapy to decrease the risk of adverse events. Finally, the Institute for Healthcare Improvement’s Age-Friendly Health System framework can be a useful when considering the addition of a new medication in an older adult’s treatment plan.

Acknowledgments

This material is the result of work supported with resources and the use of facilities at the New England Geriatrics Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, and the Cincinnati VeteransAffairs Medical Center.

References

1. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2018;71(19):e127-e248. doi:10.1016/j.jacc.2017.11.006

2. Davis LL. Hypertension: how low to go when treating older adults. J Nurse Pract. 2019;15(1):1-6. doi:10.1016/j.nurpra.2018.10.010

3. Qaseem A, Wilt TJ, Rich R, et al. Pharmacologic Treatment of Hypertension in Adults Aged 60 Years or Older to Higher Versus Lower Blood Pressure Targets: A Clinical Practice Guideline From the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med. 2017;166(6):430-437. doi:10.7326/M16-1785

4. Aronow WS, Fleg JL, Pepine CJ, et al. ACCF/AHA 2011 expert consensus document on hypertension in the elderly: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus documents developed in collaboration with the American Academy of Neurology, American Geriatrics Society, American Society for Preventive Cardiology, American Society of Hypertension, American Society of Nephrology, Association of Black Cardiologists, and European Society of Hypertension. J Am Coll Cardiol. 2011;57(20):2037-2114. doi:10.1016/j.jacc.2011.01.008

5. Makam AN, Boscardin WJ, Miao Y, Steinman MA. Risk of thiazide-induced metabolic adverse events in older adults. J Am Geriatr Soc. 2014;62(6):1039-1045. doi:10.1111/jgs.12839

6. Chow KM, Szeto CC, Wong TY, Leung CB, Li PK. Risk factors for thiazide-induced hyponatraemia. QJM. 2003;96(12):911-917. doi:10.1093/qjmed/hcg157

7. Clayton JA, Rodgers S, Blakey J, Avery A, Hall IP. Thiazide diuretic prescription and electrolyte abnormalities in primary care. Br J Clin Pharmacol. 2006;61(1):87-95. doi:10.1111/j.1365-2125.2005.02531.x

8. Shah BM, Hajjar ER. Polypharmacy, adverse drug reactions, and geriatric syndromes. Clin Geriatr Med. 2012;28(2):173-186. doi:10.1016/j.cger.2012.01.002

9. Benetos A, Petrovic M, Strandberg T. Hypertension management in older and frail older patients. Circ Res. 2019;124(7):1045-1060. doi:10.1161/CIRCRESAHA.118.313236

10. Sherrill B, Halpern M, Khan S, Zhang J, Panjabi S. Single-pill vs free-equivalent combination therapies for hypertension: a meta-analysis of health care costs and adherence. J Clin Hypertens (Greenwich). 2011;13(12):898-909. doi:10.1111/j.1751-7176.2011.00550.x

11. Mate K, Fulmer T, Pelton L, et al. Evidence for the 4Ms: interactions and outcomes across the care continuum. J Aging Health. 2021;33(7-8):469-481. doi:10.1177/0898264321991658

References

1. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol. 2018;71(19):e127-e248. doi:10.1016/j.jacc.2017.11.006

2. Davis LL. Hypertension: how low to go when treating older adults. J Nurse Pract. 2019;15(1):1-6. doi:10.1016/j.nurpra.2018.10.010

3. Qaseem A, Wilt TJ, Rich R, et al. Pharmacologic Treatment of Hypertension in Adults Aged 60 Years or Older to Higher Versus Lower Blood Pressure Targets: A Clinical Practice Guideline From the American College of Physicians and the American Academy of Family Physicians. Ann Intern Med. 2017;166(6):430-437. doi:10.7326/M16-1785

4. Aronow WS, Fleg JL, Pepine CJ, et al. ACCF/AHA 2011 expert consensus document on hypertension in the elderly: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus documents developed in collaboration with the American Academy of Neurology, American Geriatrics Society, American Society for Preventive Cardiology, American Society of Hypertension, American Society of Nephrology, Association of Black Cardiologists, and European Society of Hypertension. J Am Coll Cardiol. 2011;57(20):2037-2114. doi:10.1016/j.jacc.2011.01.008

5. Makam AN, Boscardin WJ, Miao Y, Steinman MA. Risk of thiazide-induced metabolic adverse events in older adults. J Am Geriatr Soc. 2014;62(6):1039-1045. doi:10.1111/jgs.12839

6. Chow KM, Szeto CC, Wong TY, Leung CB, Li PK. Risk factors for thiazide-induced hyponatraemia. QJM. 2003;96(12):911-917. doi:10.1093/qjmed/hcg157

7. Clayton JA, Rodgers S, Blakey J, Avery A, Hall IP. Thiazide diuretic prescription and electrolyte abnormalities in primary care. Br J Clin Pharmacol. 2006;61(1):87-95. doi:10.1111/j.1365-2125.2005.02531.x

8. Shah BM, Hajjar ER. Polypharmacy, adverse drug reactions, and geriatric syndromes. Clin Geriatr Med. 2012;28(2):173-186. doi:10.1016/j.cger.2012.01.002

9. Benetos A, Petrovic M, Strandberg T. Hypertension management in older and frail older patients. Circ Res. 2019;124(7):1045-1060. doi:10.1161/CIRCRESAHA.118.313236

10. Sherrill B, Halpern M, Khan S, Zhang J, Panjabi S. Single-pill vs free-equivalent combination therapies for hypertension: a meta-analysis of health care costs and adherence. J Clin Hypertens (Greenwich). 2011;13(12):898-909. doi:10.1111/j.1751-7176.2011.00550.x

11. Mate K, Fulmer T, Pelton L, et al. Evidence for the 4Ms: interactions and outcomes across the care continuum. J Aging Health. 2021;33(7-8):469-481. doi:10.1177/0898264321991658

Issue
Federal Practitioner - 41(2)a
Issue
Federal Practitioner - 41(2)a
Page Number
58
Page Number
58
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>0224 FED Hyponatremia</fileName> <TBEID>0C02EFC9.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02EFC9</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240201T115718</firstPublished> <LastPublished>20240201T115719</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240201T115718</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Stephanie Pagliuca, MDa,b; Caroline Wagner, PharmDc; Brittany L. Pietruszka, PharmDc; Shivani K. Jindal, MD, MPHb,c,d</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>Hypertension is a major risk factor for heart disease, stroke, and kidney disease. The prevalence of hypertension increases with age, primarily due to age-relat</metaDescription> <articlePDF/> <teaserImage/> <title>Thiazide-Induced Hyponatremia Presenting as a Fall in an Older Adult</title> <deck/> <eyebrow>Case in Point</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>February</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>2</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2953</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>February 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Case in Point | 2953<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">45</term> <term>112</term> </sections> <topics> <term canonical="true">194</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Thiazide-Induced Hyponatremia Presenting as a Fall in an Older Adult</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Regardless of age, first-line therapy for uncomplicated hypertension includes thiazide diuretics, long-acting calcium channel blockers, and renin-angiotensin system inhibitors. Even though older adults are often at increased risk of adverse drug events, specific guidelines for choosing between different classes of antihypertensives are lacking. Given the prevalence of hypertension in older adults, clinicians should be aware of the increased risk of electrolyte disorders after the initiation of thiazide diuretics in this population. <br/><br/><b>Case Presentation:</b> A patient aged &gt; 90 years fell getting out of his bed 2 weeks following initiation of hydrochlorothiazide 25 mg daily medication therapy. Laboratory tests revealed a urine sodium of 35 mmol/L most consistent with hypovolemic hypoosmotic hyponatremia secondary to thiazide initiation. Hydrochlorothiazide was discontinued and sodium gradually normalized over the next 2 weeks without any other intervention.<br/><br/><b>Conclusions:</b> Despite being recommended as first-line therapy for uncomplicated hypertension, thiazide diuretics may cause more harm than good in older adults with risk factors for thiazide-induced hyponatremia, which should be considered before initiation.</p> <p><span class="Drop">H</span>ypertension is a major risk factor for heart disease, stroke, and kidney disease.<sup> </sup>The prevalence of hypertension increases with age, primarily due to age-related changes in arterial physiology.<sup>1</sup> For older adults, current guidelines regarding blood pressure (BP) treatment goals vary. The American Heart Association/American College of Cardiology 2017 clinical practice guidelines recommend a systolic BP (SBP) treatment goal of &lt; 130 mm Hg for community-dwelling, ambulatory, noninstitutionalized adults aged ≥ 65 years; whereas the American College of Physicians/American Academy of Family Physicians recommend a goal of &lt; 150 mm Hg for those aged ≥ 60 years without comorbidities and &lt; 140 mm Hg for those with increased cardiovascular risk.<sup>1-3</sup> Regardless of the specific threshold, agreement that some degree of BP control even in those with advanced age improves outcomes.<sup>2</sup> </p> <p>First-line therapy for uncomplicated hypertension includes thiazide diuretics, long-acting calcium channel blockers, and renin-angiotensin system inhibitors. When choosing between these options, it is recommended to engage in shared decision making and to consider the patient’s comorbidities. Among patients who are likely to require a second agent (eg, if initial BP is &gt; 20/10 mm Hg above goal), it is recommended to begin both drugs at the same time, preferably benazepril plus amlodipine due to the reduction in cardiovascular events reported in the ACCOMPLISH trial.<sup>4</sup> If BP remains elevated despite 2 agents at moderate to maximum doses, it is important to investigate for secondary hypertension causes and to explore medication adherence as possible etiologies of treatment failure. Older adults are often at higher risk of adverse drug events due to age-related changes in pharmacodynamics. Despite this, there are no guidelines for choosing between different classes of antihypertensives in this population. We present a case of thiazide-induced hyponatremia in an older adult and review the risks of thiazide use in this population. </p> <h2>Case Presentation</h2> <p>A man aged &gt; 90 years was admitted to the hospital after a syncopal episode. His history was significant for hypertension, hyperlipidemia, and vitamin D deficiency. At the time, his home medications were amlodipine 5 mg daily, atorvastatin 40 mg daily, ergocalciferol 50,000 IU weekly, and polyethylene glycol 17 g daily as needed. His syncope workup was unremarkable and included negative orthostatic vital signs, normal serial troponins, an electrocardiogram without ischemic changes, normal serum creatinine, sodium, and glucose, and a head computed tomography without any acute abnormality. Throughout the patient’s hospital stay, he had multiple elevated SBP readings, including many &gt; 200 mm Hg. On discharge, in addition to continuing his home medications, he was started on valsartan 20 mg daily and enrolled in a remote BP monitoring program. </p> <p>Three weeks later, the patient was seen by their primary care practitioner for follow-up. He reported adherence to his antihypertensive regimen. However, his remote BP monitoring revealed persistently elevated BPs, with an average of 179/79 mm Hg, a high of 205/85 mm Hg, and a low of 150/67 mm Hg over the previous 7 days. Laboratory tests obtained at the visit were notable for serum sodium of 138 mmol/L and potassium of 4.1 mmol/L. His weight was 87 kg. Given persistently elevated BP readings, in addition to continuing his amlodipine 5 mg daily and valsartan 20 mg daily, he was started on hydrochlorothiazide 25 mg daily, with plans to repeat a basic metabolic panel in 2 weeks.Two weeks later, he fell after getting out of his bed. On examination, he was noted to have dry mucous membranes, and although no formal delirium screening was performed, he was able to repeat the months of the year backward. Vital signs were notable for positive postural hypertension, and his laboratory tests revealed a normal serum creatinine, serum sodium of 117 mmol/L <hl name="33671"/>(reference range, 135-145 mmol/L), serum potassium of 3.2 mmol/L (reference range, 3.0-5.0 mmol/L), a low serum osmolality, and urine sodium of 35 mmol/L<b> </b>most consistent with hypovolemic hypoosmotic hyponatremia secondary to thiazide initiation. The patient’s hydrochlorothiazide was discontinued, and he was admitted to the hospital for close monitoring. His sodium levels gradually normalized over the next 2 weeks without any other intervention.</p> <h2>Discussion</h2> <p>Although thiazide diuretics are recommended as first-line therapy for uncomplicated hypertension, they are known to cause electrolyte abnormalities, including hypomagnesemia, hypokalemia, and hyponatremia.<sup>4</sup> These metabolic derangements are more likely to occur in older adults. One study of adults aged ≥ 65 years found that at 9 months of follow-up, 14.3% of new thiazide users had developed a thiazide-related metabolic adverse event (hyponatremia &lt; 135 mmol/L, hypokalemia &lt; 3.5 mmol/L, and decrease in estimated glomerular filtration rate by &gt; 25%) compared with 6.0% of nonusers (<i>P</i> &lt; .001; number needed to harm [NNH] = 12).<sup>5</sup> In addition, 3.8% of new thiazide users had an emergency department visit or were hospitalized for complications related to thiazides compared with only 2.0% of nonusers (<i>P</i> = .02; NNH = 56).<sup>5</sup> Independent risk factors for thiazide-induced hyponatremia include high-comorbidity burden, low body weight, low-normal or unmeasured serum sodium, low potassium, and aged &gt; 70 years.<sup>5-7</sup> Each 10-year increment in age is associated with a 2-fold increase in risk, suggesting that older adults are at a much higher risk for hyponatremia than their younger peers.<sup>6</sup></p> <p>Despite their designation as a first-line option for uncomplicated hypertension, thiazide diuretics may cause more harm than good in some older adults, especially those with additional risk factors for thiazide-induced hyponatremia. In this population, these adverse effects should be discussed before starting thiazides for the treatment of hypertension. If thiazides are initiated, they should be started at the lowest possible dose, and plans made to monitor bloodwork within 1 to 2 weeks of initiation or dose change and periodically thereafter while the patient remains on the therapy.</p> <h3>Medication Management in Older Adults </h3> <p>Due to the risks of medication use in older adults, the phrase “start low, go slow” is commonly used in geriatric medicine to describe the optimal method for initiation and up-titration of new medication with the hope of mitigating adverse drug events. In our case, we started valsartan at 20 mg daily—one-fourth the recommended initial dose. Although this strategy is reasonable to “start low,” we were not surprised to find that the patient’s BP did not markedly improve on such a low dose. The team could have increased the valsartan dose to a therapeutically efficacious dose before choosing to add another hypertensive agent. In alignment with geriatric prescribing principles, starting at the lowest possible dose of hydrochlorothiazide is recommended.<sup>5</sup> However, the clinician started hydrochlorothiazide at 25 mg daily, potentially increasing this patient’s risk of electrolyte abnormalities and eventual fall. </p> <p>Managing hypertension also invites a discussion of polypharmacy and medication adherence. Older adults are at risk of polypharmacy, defined as the prescription of 5 or more medications.<sup>8</sup> Polypharmacy is associated with increased hospitalizations, higher costs of care for individuals and health care systems, increased risks of adverse drug events, medication nonadherence, and lower quality of life for patients.<sup>9</sup> In some situations, the risks of polypharmacy may outweigh the benefits of using multiple antihypertensives with different mechanisms of action if patients can reach their BP goal on the maximum dose of a single agent. For patients taking multiple antihypertensives, it is important to routinely monitor BP and assess whether deprescribing is indicated. Cognitive impairment and decreased social support may affect medication adherence for older adults.<sup>6</sup> Clinicians should be aware of strategies, such as medication reminders and pillboxes, to increase antihypertensive medication adherence. Polypills that contain 2 antihypertensives can be another tool used to manage older adults to increase adherence and decrease health care costs.<sup>10<br/><br/></sup>A current strategy that encompasses discussing many, if not all, of these noted elements is the Institute for Healthcare Improvement’s Age-Friendly Health System. This framework uses evidence-based tools to provide care for older adults across all clinical settings and highlights the 4Ms: what matters, medication, mentation, and mobility.<sup>11</sup> Medication considers whether a medication is necessary, whether its use has benefits that outweigh the risks, and how it interacts with what matters, mentation, and mobility. In particular, what matters plays an important role in hypertension management in older adults given the recommended target BP differs, depending on which specialty organization guideline is followed. By better understanding what matters to patients, including their goals and priorities, clinicians can engage patients in shared decision making and provide individualized recommendations based on geriatric principles (eg, start low, go slow, principles of medication adherence) and patient comorbidities (eg, medical history and risk factors for hyponatremia) to help patients make a more informed choice about their antihypertensive treatment regimen (Figure). </p> <h2>Conclusions</h2> <p>This case illustrates the need for a specialized approach to hypertension management in older adults and the risks of thiazide diuretics in this population. Clinicians should consider BP goals, patient-specific factors, and principles of medication management in older adults. If initiating thiazide therapy, discuss the risks associated with use, start at the lowest possible dose, and monitor bloodwork within 1 to 2 weeks of initiation/dose change and periodically thereafter while the patient remains on the therapy to decrease the risk of adverse events. Finally, the Institute for Healthcare Improvement’s Age-Friendly Health System framework can be a useful when considering the addition of a new medication in an older adult’s treatment plan.</p> <h3> Acknowledgments </h3> <p> <em>This material is the result of work supported with resources and the use of facilities at the New England Geriatrics Research, Education, and Clinical Center, Veterans Affairs Boston Healthcare System, and the Cincinnati VeteransAffairs Medical Center.</em> </p> <h3> Author affiliations </h3> <p> <em><sup>a</sup>New England Geriatrics Research, Education, and Clinical Center (GRECC), Veterans Affairs Boston Healthcare System, Massachusetts<br/><br/><sup>b</sup>Boston University Chobanian &amp; Avedisian School of Medicine, Massachusetts<sup>c</sup>Veterans Affairs Boston Healthcare System, Massachusetts<br/><br/><sup>d</sup>Cincinnati Veterans Affairs Medical Center, Ohio</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 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>This manuscript has been reviewed by the Veterans Affairs Boston Privacy Office prior to submission. We have received written consent and release of information from this veteran to use details of their case for this manuscript.</em> </p> <h3> References </h3> <p class="reference"> 1. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation and Management of High Blood Pressure in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines.<i> J Am Coll Cardiol</i>. 2018;71(19):e127-e248. doi:10.1016/j.jacc.2017.11.006<br/><br/> 2. Davis LL. Hypertension: how low to go when treating older adults. <i>J Nurse Pract</i>. 2019;15(1):1-6. doi:10.1016/j.nurpra.2018.10.010<br/><br/> 3. Qaseem A, Wilt TJ, Rich R, et al. Pharmacologic Treatment of Hypertension in Adults Aged 60 Years or Older to Higher Versus Lower Blood Pressure Targets: A Clinical Practice Guideline From the American College of Physicians and the American Academy of Family Physicians. <i>Ann Intern Med</i>. 2017;166(6):430-437. doi:10.7326/M16-1785<br/><br/> 4. Aronow WS, Fleg JL, Pepine CJ, et al. ACCF/AHA 2011 expert consensus document on hypertension in the elderly: a report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus documents developed in collaboration with the American Academy of Neurology, American Geriatrics Society, American Society for Preventive Cardiology, American Society of Hypertension, American Society of Nephrology, Association of Black Cardiologists, and European Society of Hypertension<i>. J Am Coll Cardiol</i>. 2011;57(20):2037-2114. doi:10.1016/j.jacc.2011.01.008<br/><br/> 5. Makam AN, Boscardin WJ, Miao Y, Steinman MA. Risk of thiazide-induced metabolic adverse events in older adults<i>. J Am Geriatr Soc</i>. 2014;62(6):1039-1045. doi:10.1111/jgs.12839<br/><br/> 6. Chow KM, Szeto CC, Wong TY, Leung CB, Li PK. Risk factors for thiazide-induced hyponatraemia. <i>QJM</i>. 2003;96(12):911-917. doi:10.1093/qjmed/hcg157<br/><br/> 7. Clayton JA, Rodgers S, Blakey J, Avery A, Hall IP. Thiazide diuretic prescription and electrolyte abnormalities in primary care. <i>Br J Clin Pharmacol</i>. 2006;61(1):87-95. doi:10.1111/j.1365-2125.2005.02531.x<br/><br/> 8. Shah BM, Hajjar ER. Polypharmacy, adverse drug reactions, and geriatric syndromes. <i>Clin Geriatr Med</i>. 2012;28(2):173-186. doi:10.1016/j.cger.2012.01.002<br/><br/> 9. Benetos A, Petrovic M, Strandberg T. Hypertension management in older and frail older patients. <i>Circ Res</i>. 2019;124(7):1045-1060. doi:10.1161/CIRCRESAHA.118.313236<br/><br/>10. Sherrill B, Halpern M, Khan S, Zhang J, Panjabi S. Single-pill vs free-equivalent combination therapies for hypertension: a meta-analysis of health care costs and adherence. <i>J Clin Hypertens (Greenwich)</i>. 2011;13(12):898-909. doi:10.1111/j.1751-7176.2011.00550.x<br/><br/>11. Mate K, Fulmer T, Pelton L, et al. Evidence for the 4Ms: interactions and outcomes across the care continuum. <i>J Aging Health</i>. 2021;33(7-8):469-481. doi:10.1177/0898264321991658</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

51-year-old woman • history of Graves disease • general fatigue, palpitations, and hand tremors • Dx?

Article Type
Changed
Wed, 11/15/2023 - 10:52
Display Headline
51-year-old woman • history of Graves disease • general fatigue, palpitations, and hand tremors • Dx?

THE CASE

A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care.

Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases.

Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.

Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level < 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).

Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.

THE DIAGNOSIS

Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole regularly until 2 months prior to presentation, when she started taking it because of worsening symptoms. She had hesitated to report her social and medication status due to embarrassment.

In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made.

Continue to: DISCUSSION

 

 

DISCUSSION

Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.1 The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.2 Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.2

Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.3,4 It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.5 Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.5

Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-­colony stimulating factor (G-CSF) therapy.5

Our patient was hospitalized, and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF was started. Meropenem 3 g/d also was administered for neutropenic fever.

The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,5 we decided to routinely monitor this patient due to the possibility of drug cross-reactivity.

Continue to: Despite repeated explanations...

 

 

Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur.

Immediate problem solved, but what about the future?

During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future.

Conversations about medication adherence. In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.6 To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.

Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.

Patients may be reluctant to provide the details of medication adherence.7 Although it is common for patients to need services for socioeconomic issues,8 health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.9 Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.10,11

Our patient received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed.

Continue to: The patient was discharged...

 

 

The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.

THE TAKEAWAY

Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events.

ACKNOWLEDGMENT
We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.

CORRESPONDENCE
Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; t.maejima.2019@gmail.com

References

1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis. Thyroid. 2004;14:459-462. doi: 10.1089/105072504323150787

2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. Drugs R D. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1

3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. Thyroid. 2009;19:559-563. doi: 10.1089/thy.2008.0364

4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. Endocr J. 2007;54:39-43. doi: 10.1507/endocrj.k05-068

5. Burch HB, Cooper DS. Management of Graves disease: a review. J Am Med Assoc. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535

6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. Eur Thyroid J. 2014;3:245-251. doi: https://doi.org/10.1159/000367990

7. Kini V, Ho PM. Interventions to improve medication adherence: a review. J Am Med Assoc. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271

8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. Int J Med Inform. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008

9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? Patient Educ Couns. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011

10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/

11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. Can Med Educ J. 2019;10:e61-e71. doi: 10.36834/cmej.61709

Article PDF
Author and Disclosure Information

Department of General Medicine and Primary Care, University of Tsukuba Hospital, Ibaraki (Dr. Maejima); Department of Medical Education Studies, International Research Center for Medical Education, Graduate School of Medicine, The University of Tokyo (Dr. Mizumoto); Department of Internal Medicine, Tokatsu Hospital, Chiba (Dr. Ito); Department of Endocrinology and Metabolism, TMG Asaka Medical Center, Saimata (Dr. Suga), Japan
t.maejima.2019@gmail.com

The authors reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 72(9)
Publications
Topics
Page Number
E8-E10
Sections
Author and Disclosure Information

Department of General Medicine and Primary Care, University of Tsukuba Hospital, Ibaraki (Dr. Maejima); Department of Medical Education Studies, International Research Center for Medical Education, Graduate School of Medicine, The University of Tokyo (Dr. Mizumoto); Department of Internal Medicine, Tokatsu Hospital, Chiba (Dr. Ito); Department of Endocrinology and Metabolism, TMG Asaka Medical Center, Saimata (Dr. Suga), Japan
t.maejima.2019@gmail.com

The authors reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

Department of General Medicine and Primary Care, University of Tsukuba Hospital, Ibaraki (Dr. Maejima); Department of Medical Education Studies, International Research Center for Medical Education, Graduate School of Medicine, The University of Tokyo (Dr. Mizumoto); Department of Internal Medicine, Tokatsu Hospital, Chiba (Dr. Ito); Department of Endocrinology and Metabolism, TMG Asaka Medical Center, Saimata (Dr. Suga), Japan
t.maejima.2019@gmail.com

The authors reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF

THE CASE

A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care.

Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases.

Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.

Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level < 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).

Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.

THE DIAGNOSIS

Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole regularly until 2 months prior to presentation, when she started taking it because of worsening symptoms. She had hesitated to report her social and medication status due to embarrassment.

In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made.

Continue to: DISCUSSION

 

 

DISCUSSION

Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.1 The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.2 Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.2

Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.3,4 It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.5 Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.5

Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-­colony stimulating factor (G-CSF) therapy.5

Our patient was hospitalized, and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF was started. Meropenem 3 g/d also was administered for neutropenic fever.

The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,5 we decided to routinely monitor this patient due to the possibility of drug cross-reactivity.

Continue to: Despite repeated explanations...

 

 

Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur.

Immediate problem solved, but what about the future?

During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future.

Conversations about medication adherence. In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.6 To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.

Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.

Patients may be reluctant to provide the details of medication adherence.7 Although it is common for patients to need services for socioeconomic issues,8 health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.9 Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.10,11

Our patient received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed.

Continue to: The patient was discharged...

 

 

The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.

THE TAKEAWAY

Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events.

ACKNOWLEDGMENT
We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.

CORRESPONDENCE
Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; t.maejima.2019@gmail.com

THE CASE

A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care.

Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases.

Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.

Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level < 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).

Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.

THE DIAGNOSIS

Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole regularly until 2 months prior to presentation, when she started taking it because of worsening symptoms. She had hesitated to report her social and medication status due to embarrassment.

In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made.

Continue to: DISCUSSION

 

 

DISCUSSION

Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.1 The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.2 Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.2

Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.3,4 It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.5 Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.5

Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-­colony stimulating factor (G-CSF) therapy.5

Our patient was hospitalized, and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF was started. Meropenem 3 g/d also was administered for neutropenic fever.

The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,5 we decided to routinely monitor this patient due to the possibility of drug cross-reactivity.

Continue to: Despite repeated explanations...

 

 

Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur.

Immediate problem solved, but what about the future?

During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future.

Conversations about medication adherence. In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.6 To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.

Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.

Patients may be reluctant to provide the details of medication adherence.7 Although it is common for patients to need services for socioeconomic issues,8 health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.9 Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.10,11

Our patient received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed.

Continue to: The patient was discharged...

 

 

The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.

THE TAKEAWAY

Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events.

ACKNOWLEDGMENT
We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.

CORRESPONDENCE
Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; t.maejima.2019@gmail.com

References

1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis. Thyroid. 2004;14:459-462. doi: 10.1089/105072504323150787

2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. Drugs R D. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1

3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. Thyroid. 2009;19:559-563. doi: 10.1089/thy.2008.0364

4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. Endocr J. 2007;54:39-43. doi: 10.1507/endocrj.k05-068

5. Burch HB, Cooper DS. Management of Graves disease: a review. J Am Med Assoc. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535

6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. Eur Thyroid J. 2014;3:245-251. doi: https://doi.org/10.1159/000367990

7. Kini V, Ho PM. Interventions to improve medication adherence: a review. J Am Med Assoc. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271

8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. Int J Med Inform. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008

9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? Patient Educ Couns. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011

10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/

11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. Can Med Educ J. 2019;10:e61-e71. doi: 10.36834/cmej.61709

References

1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis. Thyroid. 2004;14:459-462. doi: 10.1089/105072504323150787

2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. Drugs R D. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1

3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. Thyroid. 2009;19:559-563. doi: 10.1089/thy.2008.0364

4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. Endocr J. 2007;54:39-43. doi: 10.1507/endocrj.k05-068

5. Burch HB, Cooper DS. Management of Graves disease: a review. J Am Med Assoc. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535

6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. Eur Thyroid J. 2014;3:245-251. doi: https://doi.org/10.1159/000367990

7. Kini V, Ho PM. Interventions to improve medication adherence: a review. J Am Med Assoc. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271

8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. Int J Med Inform. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008

9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? Patient Educ Couns. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011

10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/

11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. Can Med Educ J. 2019;10:e61-e71. doi: 10.36834/cmej.61709

Issue
The Journal of Family Practice - 72(9)
Issue
The Journal of Family Practice - 72(9)
Page Number
E8-E10
Page Number
E8-E10
Publications
Publications
Topics
Article Type
Display Headline
51-year-old woman • history of Graves disease • general fatigue, palpitations, and hand tremors • Dx?
Display Headline
51-year-old woman • history of Graves disease • general fatigue, palpitations, and hand tremors • Dx?
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>JFP1123_CROnline</fileName> <TBEID>0C02E920.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02E920</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>51-year-old woman • history of G</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-JFP</TBLocation> <QCDate/> <firstPublished>20231115T094707</firstPublished> <LastPublished>20231115T094707</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20231115T094706</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Takuya Maejima, MD; Junki Mizumoto, MD; Yoshiko Ito, MD</byline> <bylineText/> <bylineFull>Takuya Maejima, MD; Junki Mizumoto, MD; Yoshiko Ito, MD</bylineFull> <bylineTitleText>We thank Jane Charbonneau, DVM, from Edanz for doing an English-language review of a draft of this manuscript.Takuya Maejima, MD, Department of General Medicine and Primary Care, University of Tsukuba Hospital, 2-1-1 Amakubo, Tsukuba, Ibaraki 305-8576 Japan; t.maejima.2019@gmail. com</bylineTitleText> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E8-E10</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitati</metaDescription> <articlePDF>299192</articlePDF> <teaserImage/> <title/> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2023</pubPubdateYear> <pubPubdateMonth>November</pubPubdateMonth> <pubPubdateDay/> <pubVolume>72</pubVolume> <pubNumber>9</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>3183</CMSID> </CMSIDs> <keywords> <keyword>endocrinology</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>jfp</publicationCode> <pubIssueName>November 2023</pubIssueName> <pubArticleType>Case Studies | 3183</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> </publicationData> <publicationData> <publicationCode>mdfam</publicationCode> <pubIssueName/> <pubArticleType/> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle/> <journalFullTitle/> <copyrightStatement/> </publicationData> </publications_g> <publications> <term canonical="true">30</term> <term>51948</term> </publications> <sections> <term canonical="true">45</term> </sections> <topics> <term>206</term> <term canonical="true">27442</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002638.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <h3>THE CASE</h3> <p>A 51-year-old Japanese woman presented with fever, sore throat, and dyspnea of less than 1 day’s duration. Although she had developed general fatigue, palpitations, and tremors of the hands 2 months earlier, she had not sought medical care. </p> <p>Her medical history included Graves disease, which had been diagnosed 13 years earlier. She reported that her only medication was methimazole 10 mg/d. She did not have any family history of endocrinopathies or hematologic diseases. <br/><br/>Physical examination revealed a body temperature of 99.7 °F; heart rate, 130 beats/min; blood pressure, 182/62 mm Hg; respiratory rate, 46 breaths/min; and oxygen saturation, 100% on room air. Pharyngeal erythema was seen. Lung sounds were clear. The patient had tremors in her hands, tenderness of the thyroid gland, and exophthalmos. No leg edema or jugular vein distension was seen.<br/><br/>Laboratory tests indicated hyperthyroidism, with a thyroid-stimulating hormone level &lt; 0.01 µIU/mL (normal range, 0.5-5 µIU/mL); free T3 level, 4.87 pg/mL (normal range, 2.3-4.3 pg/mL); and free T4 level, 2.97 ng/dL (normal range, 0.9-1.7 ng/dL). The patient also had a white blood cell (WBC) count of 1020 cells/µL (normal range, 3500-9000 cells/µL) and neutrophil count of 5 cells/µL (normal range, 1500-6500 cells/µL).<br/><br/>Other blood cell counts were normal, and a chest x-ray did not reveal any abnormal findings. In addition, there was no evidence to suggest hematologic malignancies or congenital neutropenia.</p> <h3>THE DIAGNOSIS</h3> <p>Based on the patient’s low WBC and neutrophil counts, agranulocytosis due to antithyroid drug therapy was suspected; however, this diagnosis would be highly unusual in the context of a 13-year history of therapy. Further history taking revealed that, because of her lack of financial means, unstable living conditions, and lack of understanding of the necessity for medication adherence, the patient had not taken methimazole <hl name="5"/>regularly until 2 months prior to presentation, when she started taking it because of worsening symptoms. She had hesitated to report her social and medication status due to embarrassment.</p> <p><hl name="6"/>In consideration of these factors, a diagnosis of exacerbation of hyperthyroidism and agranulocytosis (due to methimazole restart and upper respiratory infection) was made. </p> <h3>DISCUSSION</h3> <p>Agranulocytosis is a severe adverse event of antithyroid agents and requires prompt diagnosis and treatment. In a 26-year study at one clinic, it occurred in approximately 0.4% of patients taking antithyroid agents.<sup>1</sup> The possible mechanisms of agranulocytosis are the direct toxicity of drugs and immune-mediated responses.<sup>2</sup> Older age, female sex, and some HLA genotypes are reported to be associated with susceptibility to agranulocytosis.<sup>2</sup> </p> <p>Although the development of agranulocytosis tends to be dose related, a small dose of antithyroid agent can sometimes cause the condition.<sup>3,4</sup> It usually occurs within the first 3 months of treatment initiation, but occasionally patients develop agranulocytosis after long-term therapy.<sup>5</sup> Interruption and subsequent resumption of the same antithyroid drug treatment also can be a risk factor for agranulocytosis, as in this case.<sup>5</sup> <br/><br/>Treatment includes drug cessation, administration of broad-spectrum antibiotics if infection is suspected, and granulocyte-­colony stimulating factor (G-CSF) therapy.<sup>5</sup> <br/><br/><span class="dingbat3">❚</span><span class="intro"> Our patient was hospitalized,</span> and methimazole was stopped immediately. Administration of potassium iodide 50 mg/d and G-CSF<b> </b>was started. Meropenem 3 g/d also was administered for neutropenic fever. <br/><br/>The patient’s condition improved, and her WBC count increased to 1640 cells/µL on Day 8 and 10,890 cells/µL on Day 9. G-CSF was stopped on Day 12 and meropenem on Day 13. Bone marrow aspiration was not performed because of improvement in lab values and her overall condition. Although monitoring of WBC count during methimazole therapy is controversial,<sup>5</sup> we decided to routinely monitor this patient due to the possibility of drug cross-reactivity. <br/><br/>Despite repeated explanations that it was dangerous for a patient who had developed agranulocytosis to take another antithyroid medication, the patient refused surgical treatment or radioiodine ablation because of her financial situation. (While all Japanese citizens are covered by a national health insurance program, patients ages 6 to 70 years are required to pay approximately 30% of medical and pharmaceutical costs.) On Day 21, potassium iodide was stopped, and propylthiouracil 300 mg/d was administered with careful follow-up. Agranulocytosis did not recur. </p> <p class="sub1">Immediate problem solved,but what about the future?</p> <p>During her hospital stay, the medical team spoke with the patient many times, during which she expressed anxiety about her health conditions and the difficulties that she had experienced in her life. The clinicians acknowledged her concerns and assured the patient of their continuing commitment to her well-being even after discharge. The patient also was advised that she should take her medication as prescribed and that if she had a fever or sore throat, she should stop the medication and seek medical care as soon as possible. The patient accepted the medical team’s advice and expressed hope for the future. </p> <p><span class="dingbat3">❚</span><span class="intro"> Conversations about medication adherence.</span> In 1 survey, about 60% of patients taking antithyroid drugs were unfamiliar with the symptoms of agranulocytosis.<sup>6</sup> To deliver safe and effective treatment and detect conditions such as agranulocytosis at an early stage, clinicians must communicate clearly with patients who have hyperthyroidism, providing sufficient explanation and ensuring understanding on the patient’s part.<br/><br/>Patients may be reluctant to provide the details of medication adherence.<sup>7</sup> Although it is common for patients to need services for socioeconomic issues,<sup>8</sup> health care professionals sometimes fail to adequately discuss these issues with patients, especially if the patients are marginalized and/or have lower economic status.<sup>9</sup> Cases such as ours underscore the importance of improving clinicians’ awareness and sensitivity to patients’ socioeconomic challenges.<sup>10,11</sup> <br/><br/><span class="dingbat3">❚</span><span class="intro"> Our patient </span>received information about welfare and other government services from a medical social worker during her hospital stay. She also was informed that she could seek assistance from medical social workers in the future if needed. <b> <br/><br/></b>The patient was discharged on Day 28. After discharge, she took propylthiouracil as prescribed (300 mg/d), and her Graves disease was well controlled. Outpatient follow-up visits were performed every 1 or 2 months. No adverse events of propylthiouracil were seen in the ensuing time.</p> <h3>THE TAKEAWAY</h3> <p>Patients with chronic conditions sometimes discontinue medications, and they may not talk about it with their medical team, especially if they have socioeconomic or other difficulties in their lives. Clinicians should consider medication nonadherence and its risk factors when patients with chronic conditions develop unexpected adverse events. <span class="end">JFP</span></p> <p class="sub5"> <caps>Acknowledgment</caps> </p> <p class="sub5">CORRESPONDENCE </p> <p class="reference"> 1. Tajiri J, Noguchi S. Antithyroid drug-induced agranulocytosis: special reference to normal white blood cell count agranulocytosis.<b> </b><i>Thyroid</i>. 2004;14:459-462. doi: 10.1089/105072504323150787<br/><br/> 2. Vicente N, Cardoso L, Barros L, et al. Antithyroid drug-induced agranulocytosis: state of the art on diagnosis and management. <i>Drugs R D</i>. 2017;17:91-96. doi: 10.1007/s40268-017-0172-1<br/><br/> 3. Takata K, Kubota S, Fukata S, et al. Methimazole-induced agranulocytosis in patients with Graves’ disease is more frequent with an initial dose of 30 mg daily than with 15 mg daily. <i>Thyroid</i>. 2009;19:559-563. doi: 10.1089/thy.2008.0364 <br/><br/> 4. Tsuboi K, Ueshiba H, Shimojo M, et al. The relation of initial methimazole dose to the incidence of methimazole-induced agranulocytosis in patients with Graves’ disease. <i>Endocr J</i>. 2007;54:39-43. doi: 10.1507/endocrj.k05-068 <br/><br/> 5. Burch HB, Cooper DS. Management of Graves disease: a review. <i>J Am Med Assoc</i>. 2015;314:2544-2554. doi: 10.1001/jama.2015.16535 <br/><br/> 6. Robinson J, Richardson M, Hickey J, et al. Patient knowledge of antithyroid drug-induced agranulocytosis. <i>Eur Thyroid J</i>. 2014;3:245-251. doi: https://doi.org/10.1159/000367990<br/><br/> 7. Kini V, Ho PM. Interventions to improve medication adherence: a review. <i>J Am Med Assoc</i>. 2018;320:2461-2473. doi: 10.1001/jama.2018.19271<br/><br/> 8. Vest JR, Grannis SJ, Haut DP, et al. Using structured and unstructured data to identify patients’ need for services that address the social determinants of health. <i>Int J Med Inform</i>. 2017;107:101-106. doi: 10.1016/j.ijmedinf.2017.09.008<br/><br/> 9. Willems S, De Maesschalck S, Deveugele M, et al. Socio-economic status of the patient and doctor-patient communication: does it make a difference? <i>Patient Educ Couns</i>. 2005;56:139-146. doi: 10.1016/j.pec.2004.02.011<br/><br/> 10. The College of Family Physicians of Canada. Best advice: social determinants of health. Accessed September 15, 2023. https://patientsmedicalhome.ca/resources/best-advice-guides/best-advice-guide-social-determinants-health/ <br/><br/> 11. Hunter K, Thomson B. A scoping review of social determinants of health curricula in post-graduate medical education. <i>Can Med Educ J</i>. 2019;10:e61-e71. doi: 10.36834/cmej.61709</p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

► History of Graves disease
► General fatigue, palpitations, and hand tremors

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
18002638.SIG
Disable zoom
Off