Commentary: Evaluating Recent BC Treatment Trials, May 2024

Article Type
Changed
Fri, 05/03/2024 - 21:47
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Support for de-escalation of axillary surgery for select patients, specifically those with cT1-2, node-negative breast cancer and a positive sentinel lymph node (SLN) biopsy, has been demonstrated in prior studies, including the ACOSOG Z0011 and AMAROS trials.[1,2] Both of these trials showed no benefit of completion axillary-node dissection (ALND) after 10 years of follow-up for these patients, and higher rates of lymphedema for ALND were observed in AMAROS. The phase 3 noninferiority SENOMAC trial aimed to validate findings from prior studies and to include groups of patients that were underrepresented (patients undergoing mastectomy, SLN with extracapsular extension, T3 tumors, and males). A total of 2540 patients with cT1-3cN0 primary breast cancer and one to two SLN macrometastases were randomly assigned to SLNB or completion ALND. The majority of patients received radiation, including nodal target volumes, as well as adjuvant systemic therapy. The estimated 5-year recurrence-free survival after SLNB only was noninferior to that seen with complete ALND (89.7%, 95% CI 87.5%-91.9%; vs 88.7%, 95% CI 86.3%-91.1%) with a hazard ratio for recurrence or death of 0.89, which was significantly (P < .001) below the noninferiority margin. These results add to the growing body of data indicating that certain patient populations can be spared more aggressive axillary surgery while maintaining excellent survival outcomes and reducing side effects.

The class of CDK 4/6 inhibitors represents a significant advance in the treatment of hormone receptor (HR)-positive breast cancer. All three CDK 4/6 inhibitors (palbociclib, abemaciclib, and ribociclib) are approved in combination with endocrine therapy in the metastatic setting. As drugs show promise in later-stage disease, they are then often studied in the curative space. Presently, abemaciclib is the only CDK 4/6 inhibitor that has been approved by the US Food and Drug Administration for the treatment of HR-positive, human epidermal growth factor receptor 2 (HER2)-negative, node-positive, high-risk early breast cancer, based on results from the monarchE trial, which demonstrated invasive disease-free survival benefit with the addition of 2 years of abemaciclib to endocrine therapy. At 4 years, the absolute difference in invasive disease-free survival (IDFS) between the groups was 6.4% (85.8% in the abemaciclib + endocrine therapy group vs 79.4% in the endocrine therapy–alone group).[3] In contrast, the PENELOPE-B and PALLAS trials did not show benefit with the addition of palbociclib to endocrine therapy in the adjuvant setting.[4,5] The phase 3 NATALEE trial randomly assigned patients with HR-positive, HER2-negative early breast cancer to ribociclib (400 mg daily for 3 weeks followed by 1 week off for 3 years) plus a nonsteroidal aromatase inhibitor (NSAI) or an NSAI alone. At the time of prespecified interim analysis, among 5101 patients, ribociclib + NSAI led to a significant improvement in IDFS compared with endocrine therapy alone (3-year IDFS was 90.4% vs 87.1%; hazard ratio 0.75; 95% CI 0.62-0.91; P = .003). It is certainly noteworthy that the trial design, endocrine therapies, and patient populations differed between these adjuvant studies; for example, NATALEE included a lower-risk population, and all patients received an NSAI (in monarchE approximately 30% received tamoxifen). The current results of NATALEE are encouraging; an absolute benefit of 3.3% should be considered and weighed against toxicities and cost, and longer follow-up is needed to further elucidate the role of ribociclib in the adjuvant space.

The meaningful impact of achieving a pathologic complete response (pCR) has been demonstrated in various prior studies. Response to neoadjuvant chemotherapy informs prognosis and helps tailor adjuvant therapy, the latter of which is particularly relevant for the HER2-positive subtype. Strategies to identify patients who are more likely to achieve pCR and predictors of early responders may aid in improving efficacy outcomes and limiting toxicities. TRAIN-3 is a single-arm, phase 2 study that included 235 and 232 patients with stage II/III HR-/HER2+ and HR+/HER2+ breast cancer, respectively, undergoing neoadjuvant chemotherapy (weekly paclitaxel D1 and D8/carboplatin AUC 6 D1/trastuzumab D1/pertuzumab D1 every 3 weeks for up to nine cycles), and was designed to evaluate radiologic and pathologic response rates and event-free survival. Response was monitored by breast MRI every 3 cycles and lymph node biopsy. Among patients with HR-/HER2+ tumors, 84 (36%; 95% CI 30-43) achieved a radiologic complete response after one to three cycles, of whom the majority (88%; 95% CI 79-94) had pCR. Patients with HR+/HER2+ tumors did not show the same degree of benefit with an MRI-based monitoring strategy; among the 138 patients (59%; 95% CI 53-66) who had a complete radiologic response after one to nine cycles, 73 (53%; 95% CI 44-61) had pCR. Additional imaging-guided modalities being studied to tailor and optimize treatment include [18F]fluorodeoxyglucose-PET-CT and volumetric MRI, in the PHERGain and I-SPY trials, respectively.[6,7]

Additional References:

  1. Giuliano AE, Ballman KV, McCall L, et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: The ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017;318:918-926. doi: 10.1001/jama.2017.11470 Source
  2. Bartels SAL, Donker M, Poncet C, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer: 10-year results of the randomized controlled EORTC 10981-22023 AMAROS trial. J Clin Oncol. 2023;41:2159-2165. doi: 10.1200/JCO.22.01565 Source
  3. Johnston SRD, Toi M, O'Shaughnessy J, et al, on behalf of the monarchE Committee Members. Abemaciclib plus endocrine therapy for hormone receptor-positive, HER2-negative, node-positive, high-risk early breast cancer (monarchE): Results from a preplanned interim analysis of a randomised, open-label, phase 3 trial. Lancet Oncol. 2023;24:77-90. doi: 10.1016/S1470-2045(22)00694-5 Source
  4. Loibl S, Marmé F, Martin M, et al. Palbociclib for residual high-risk invasive HR-positive and HER2-negative early breast cancer—The Penelope-B trial. J Clin Oncol. 2021;39:1518-1530. doi: 10.1200/JCO.20.03639 Source
  5. Gnant M, Dueck AC, Frantal S, et al, on behalf of the PALLAS groups and investigators. Adjuvant palbociclib for early breast cancer: The PALLAS trial results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol. 2022;40:282-293. doi: 10.1200/JCO.21.02554 Source
  6. Pérez-García JM, Cortés J, Ruiz-Borrego M, et al, on behalf of the PHERGain trial investigators. 3-year invasive disease-free survival with chemotherapy de-escalation using an 18F-FDG-PET-based, pathological complete response-adapted strategy in HER2-positive early breast cancer (PHERGain): A randomised, open-label, phase 2 trial. Lancet. 2024;403:1649-1659. doi: 10.1016/S0140-6736(24)00054-0 Source
  7. Hylton NM, Gatsonis CA, Rosen MA, et al, for the ACRIN 6657 trial team and I-SPY 1 trial investigators. Neoadjuvant chemotherapy for breast cancer: Functional tumor volume by MR imaging predicts recurrence-free survival-results from the ACRIN 6657/CALGB 150007 I-SPY 1 trial. Radiology. 2016;279:44-55. doi: 10.1148/radiol.2015150013 Source
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Support for de-escalation of axillary surgery for select patients, specifically those with cT1-2, node-negative breast cancer and a positive sentinel lymph node (SLN) biopsy, has been demonstrated in prior studies, including the ACOSOG Z0011 and AMAROS trials.[1,2] Both of these trials showed no benefit of completion axillary-node dissection (ALND) after 10 years of follow-up for these patients, and higher rates of lymphedema for ALND were observed in AMAROS. The phase 3 noninferiority SENOMAC trial aimed to validate findings from prior studies and to include groups of patients that were underrepresented (patients undergoing mastectomy, SLN with extracapsular extension, T3 tumors, and males). A total of 2540 patients with cT1-3cN0 primary breast cancer and one to two SLN macrometastases were randomly assigned to SLNB or completion ALND. The majority of patients received radiation, including nodal target volumes, as well as adjuvant systemic therapy. The estimated 5-year recurrence-free survival after SLNB only was noninferior to that seen with complete ALND (89.7%, 95% CI 87.5%-91.9%; vs 88.7%, 95% CI 86.3%-91.1%) with a hazard ratio for recurrence or death of 0.89, which was significantly (P < .001) below the noninferiority margin. These results add to the growing body of data indicating that certain patient populations can be spared more aggressive axillary surgery while maintaining excellent survival outcomes and reducing side effects.

The class of CDK 4/6 inhibitors represents a significant advance in the treatment of hormone receptor (HR)-positive breast cancer. All three CDK 4/6 inhibitors (palbociclib, abemaciclib, and ribociclib) are approved in combination with endocrine therapy in the metastatic setting. As drugs show promise in later-stage disease, they are then often studied in the curative space. Presently, abemaciclib is the only CDK 4/6 inhibitor that has been approved by the US Food and Drug Administration for the treatment of HR-positive, human epidermal growth factor receptor 2 (HER2)-negative, node-positive, high-risk early breast cancer, based on results from the monarchE trial, which demonstrated invasive disease-free survival benefit with the addition of 2 years of abemaciclib to endocrine therapy. At 4 years, the absolute difference in invasive disease-free survival (IDFS) between the groups was 6.4% (85.8% in the abemaciclib + endocrine therapy group vs 79.4% in the endocrine therapy–alone group).[3] In contrast, the PENELOPE-B and PALLAS trials did not show benefit with the addition of palbociclib to endocrine therapy in the adjuvant setting.[4,5] The phase 3 NATALEE trial randomly assigned patients with HR-positive, HER2-negative early breast cancer to ribociclib (400 mg daily for 3 weeks followed by 1 week off for 3 years) plus a nonsteroidal aromatase inhibitor (NSAI) or an NSAI alone. At the time of prespecified interim analysis, among 5101 patients, ribociclib + NSAI led to a significant improvement in IDFS compared with endocrine therapy alone (3-year IDFS was 90.4% vs 87.1%; hazard ratio 0.75; 95% CI 0.62-0.91; P = .003). It is certainly noteworthy that the trial design, endocrine therapies, and patient populations differed between these adjuvant studies; for example, NATALEE included a lower-risk population, and all patients received an NSAI (in monarchE approximately 30% received tamoxifen). The current results of NATALEE are encouraging; an absolute benefit of 3.3% should be considered and weighed against toxicities and cost, and longer follow-up is needed to further elucidate the role of ribociclib in the adjuvant space.

The meaningful impact of achieving a pathologic complete response (pCR) has been demonstrated in various prior studies. Response to neoadjuvant chemotherapy informs prognosis and helps tailor adjuvant therapy, the latter of which is particularly relevant for the HER2-positive subtype. Strategies to identify patients who are more likely to achieve pCR and predictors of early responders may aid in improving efficacy outcomes and limiting toxicities. TRAIN-3 is a single-arm, phase 2 study that included 235 and 232 patients with stage II/III HR-/HER2+ and HR+/HER2+ breast cancer, respectively, undergoing neoadjuvant chemotherapy (weekly paclitaxel D1 and D8/carboplatin AUC 6 D1/trastuzumab D1/pertuzumab D1 every 3 weeks for up to nine cycles), and was designed to evaluate radiologic and pathologic response rates and event-free survival. Response was monitored by breast MRI every 3 cycles and lymph node biopsy. Among patients with HR-/HER2+ tumors, 84 (36%; 95% CI 30-43) achieved a radiologic complete response after one to three cycles, of whom the majority (88%; 95% CI 79-94) had pCR. Patients with HR+/HER2+ tumors did not show the same degree of benefit with an MRI-based monitoring strategy; among the 138 patients (59%; 95% CI 53-66) who had a complete radiologic response after one to nine cycles, 73 (53%; 95% CI 44-61) had pCR. Additional imaging-guided modalities being studied to tailor and optimize treatment include [18F]fluorodeoxyglucose-PET-CT and volumetric MRI, in the PHERGain and I-SPY trials, respectively.[6,7]

Additional References:

  1. Giuliano AE, Ballman KV, McCall L, et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: The ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017;318:918-926. doi: 10.1001/jama.2017.11470 Source
  2. Bartels SAL, Donker M, Poncet C, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer: 10-year results of the randomized controlled EORTC 10981-22023 AMAROS trial. J Clin Oncol. 2023;41:2159-2165. doi: 10.1200/JCO.22.01565 Source
  3. Johnston SRD, Toi M, O'Shaughnessy J, et al, on behalf of the monarchE Committee Members. Abemaciclib plus endocrine therapy for hormone receptor-positive, HER2-negative, node-positive, high-risk early breast cancer (monarchE): Results from a preplanned interim analysis of a randomised, open-label, phase 3 trial. Lancet Oncol. 2023;24:77-90. doi: 10.1016/S1470-2045(22)00694-5 Source
  4. Loibl S, Marmé F, Martin M, et al. Palbociclib for residual high-risk invasive HR-positive and HER2-negative early breast cancer—The Penelope-B trial. J Clin Oncol. 2021;39:1518-1530. doi: 10.1200/JCO.20.03639 Source
  5. Gnant M, Dueck AC, Frantal S, et al, on behalf of the PALLAS groups and investigators. Adjuvant palbociclib for early breast cancer: The PALLAS trial results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol. 2022;40:282-293. doi: 10.1200/JCO.21.02554 Source
  6. Pérez-García JM, Cortés J, Ruiz-Borrego M, et al, on behalf of the PHERGain trial investigators. 3-year invasive disease-free survival with chemotherapy de-escalation using an 18F-FDG-PET-based, pathological complete response-adapted strategy in HER2-positive early breast cancer (PHERGain): A randomised, open-label, phase 2 trial. Lancet. 2024;403:1649-1659. doi: 10.1016/S0140-6736(24)00054-0 Source
  7. Hylton NM, Gatsonis CA, Rosen MA, et al, for the ACRIN 6657 trial team and I-SPY 1 trial investigators. Neoadjuvant chemotherapy for breast cancer: Functional tumor volume by MR imaging predicts recurrence-free survival-results from the ACRIN 6657/CALGB 150007 I-SPY 1 trial. Radiology. 2016;279:44-55. doi: 10.1148/radiol.2015150013 Source

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Support for de-escalation of axillary surgery for select patients, specifically those with cT1-2, node-negative breast cancer and a positive sentinel lymph node (SLN) biopsy, has been demonstrated in prior studies, including the ACOSOG Z0011 and AMAROS trials.[1,2] Both of these trials showed no benefit of completion axillary-node dissection (ALND) after 10 years of follow-up for these patients, and higher rates of lymphedema for ALND were observed in AMAROS. The phase 3 noninferiority SENOMAC trial aimed to validate findings from prior studies and to include groups of patients that were underrepresented (patients undergoing mastectomy, SLN with extracapsular extension, T3 tumors, and males). A total of 2540 patients with cT1-3cN0 primary breast cancer and one to two SLN macrometastases were randomly assigned to SLNB or completion ALND. The majority of patients received radiation, including nodal target volumes, as well as adjuvant systemic therapy. The estimated 5-year recurrence-free survival after SLNB only was noninferior to that seen with complete ALND (89.7%, 95% CI 87.5%-91.9%; vs 88.7%, 95% CI 86.3%-91.1%) with a hazard ratio for recurrence or death of 0.89, which was significantly (P < .001) below the noninferiority margin. These results add to the growing body of data indicating that certain patient populations can be spared more aggressive axillary surgery while maintaining excellent survival outcomes and reducing side effects.

The class of CDK 4/6 inhibitors represents a significant advance in the treatment of hormone receptor (HR)-positive breast cancer. All three CDK 4/6 inhibitors (palbociclib, abemaciclib, and ribociclib) are approved in combination with endocrine therapy in the metastatic setting. As drugs show promise in later-stage disease, they are then often studied in the curative space. Presently, abemaciclib is the only CDK 4/6 inhibitor that has been approved by the US Food and Drug Administration for the treatment of HR-positive, human epidermal growth factor receptor 2 (HER2)-negative, node-positive, high-risk early breast cancer, based on results from the monarchE trial, which demonstrated invasive disease-free survival benefit with the addition of 2 years of abemaciclib to endocrine therapy. At 4 years, the absolute difference in invasive disease-free survival (IDFS) between the groups was 6.4% (85.8% in the abemaciclib + endocrine therapy group vs 79.4% in the endocrine therapy–alone group).[3] In contrast, the PENELOPE-B and PALLAS trials did not show benefit with the addition of palbociclib to endocrine therapy in the adjuvant setting.[4,5] The phase 3 NATALEE trial randomly assigned patients with HR-positive, HER2-negative early breast cancer to ribociclib (400 mg daily for 3 weeks followed by 1 week off for 3 years) plus a nonsteroidal aromatase inhibitor (NSAI) or an NSAI alone. At the time of prespecified interim analysis, among 5101 patients, ribociclib + NSAI led to a significant improvement in IDFS compared with endocrine therapy alone (3-year IDFS was 90.4% vs 87.1%; hazard ratio 0.75; 95% CI 0.62-0.91; P = .003). It is certainly noteworthy that the trial design, endocrine therapies, and patient populations differed between these adjuvant studies; for example, NATALEE included a lower-risk population, and all patients received an NSAI (in monarchE approximately 30% received tamoxifen). The current results of NATALEE are encouraging; an absolute benefit of 3.3% should be considered and weighed against toxicities and cost, and longer follow-up is needed to further elucidate the role of ribociclib in the adjuvant space.

The meaningful impact of achieving a pathologic complete response (pCR) has been demonstrated in various prior studies. Response to neoadjuvant chemotherapy informs prognosis and helps tailor adjuvant therapy, the latter of which is particularly relevant for the HER2-positive subtype. Strategies to identify patients who are more likely to achieve pCR and predictors of early responders may aid in improving efficacy outcomes and limiting toxicities. TRAIN-3 is a single-arm, phase 2 study that included 235 and 232 patients with stage II/III HR-/HER2+ and HR+/HER2+ breast cancer, respectively, undergoing neoadjuvant chemotherapy (weekly paclitaxel D1 and D8/carboplatin AUC 6 D1/trastuzumab D1/pertuzumab D1 every 3 weeks for up to nine cycles), and was designed to evaluate radiologic and pathologic response rates and event-free survival. Response was monitored by breast MRI every 3 cycles and lymph node biopsy. Among patients with HR-/HER2+ tumors, 84 (36%; 95% CI 30-43) achieved a radiologic complete response after one to three cycles, of whom the majority (88%; 95% CI 79-94) had pCR. Patients with HR+/HER2+ tumors did not show the same degree of benefit with an MRI-based monitoring strategy; among the 138 patients (59%; 95% CI 53-66) who had a complete radiologic response after one to nine cycles, 73 (53%; 95% CI 44-61) had pCR. Additional imaging-guided modalities being studied to tailor and optimize treatment include [18F]fluorodeoxyglucose-PET-CT and volumetric MRI, in the PHERGain and I-SPY trials, respectively.[6,7]

Additional References:

  1. Giuliano AE, Ballman KV, McCall L, et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: The ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017;318:918-926. doi: 10.1001/jama.2017.11470 Source
  2. Bartels SAL, Donker M, Poncet C, et al. Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer: 10-year results of the randomized controlled EORTC 10981-22023 AMAROS trial. J Clin Oncol. 2023;41:2159-2165. doi: 10.1200/JCO.22.01565 Source
  3. Johnston SRD, Toi M, O'Shaughnessy J, et al, on behalf of the monarchE Committee Members. Abemaciclib plus endocrine therapy for hormone receptor-positive, HER2-negative, node-positive, high-risk early breast cancer (monarchE): Results from a preplanned interim analysis of a randomised, open-label, phase 3 trial. Lancet Oncol. 2023;24:77-90. doi: 10.1016/S1470-2045(22)00694-5 Source
  4. Loibl S, Marmé F, Martin M, et al. Palbociclib for residual high-risk invasive HR-positive and HER2-negative early breast cancer—The Penelope-B trial. J Clin Oncol. 2021;39:1518-1530. doi: 10.1200/JCO.20.03639 Source
  5. Gnant M, Dueck AC, Frantal S, et al, on behalf of the PALLAS groups and investigators. Adjuvant palbociclib for early breast cancer: The PALLAS trial results (ABCSG-42/AFT-05/BIG-14-03). J Clin Oncol. 2022;40:282-293. doi: 10.1200/JCO.21.02554 Source
  6. Pérez-García JM, Cortés J, Ruiz-Borrego M, et al, on behalf of the PHERGain trial investigators. 3-year invasive disease-free survival with chemotherapy de-escalation using an 18F-FDG-PET-based, pathological complete response-adapted strategy in HER2-positive early breast cancer (PHERGain): A randomised, open-label, phase 2 trial. Lancet. 2024;403:1649-1659. doi: 10.1016/S0140-6736(24)00054-0 Source
  7. Hylton NM, Gatsonis CA, Rosen MA, et al, for the ACRIN 6657 trial team and I-SPY 1 trial investigators. Neoadjuvant chemotherapy for breast cancer: Functional tumor volume by MR imaging predicts recurrence-free survival-results from the ACRIN 6657/CALGB 150007 I-SPY 1 trial. Radiology. 2016;279:44-55. doi: 10.1148/radiol.2015150013 Source
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
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

Commentary: Comparisons Among PsA Therapies, May 2024

Article Type
Changed
Fri, 04/26/2024 - 15:54
Dr. Chandran scans the journals, so you don't have to!

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Papers on psoriatic arthritis (PsA) published this month have focused on the clinical characteristics of PsA and pharmacologic treatment. Persistent inflammation leads to joint damage that is initially evident on imaging. Hen and colleagues evaluated 122 newly diagnosed, disease-modifying antirheumatic drug (DMARD)–naive patients with early PsA from the Leeds Spondyloarthropathy Register for Research and Observation cohort using conventional radiography and ultrasonography. Overall, 4655 hand and feet joints were assessed in 122 patients, of whom 24.6% had bone erosions at baseline; higher disease activity was observed in patients who did vs those who did not have bone erosions (P < .05). The prevalence of erosions was less in patients who had PsA symptoms < 8 months vs > 24 months (17.5% vs 24.3%, respectively). The agreement between conventional radiography and ultrasonography was high, with conventional radiography detecting more erosions. Thus, joint damage is seen early in patients with PsA; making a diagnosis within 8 months of symptoms is likely to lead to less joint damage and better outcomes.

 

Bimekizumab is a novel biologic therapy that inhibits interleukin (IL)–17A and IL-17F and is efficacious in the treatment of psoriasis, PsA, and axial spondyloarthritis. In the absence of a formal head-to-head study, matching-adjusted indirect comparison is a method to evaluate comparative effectiveness. Warren and colleagues ran a study that included biological DMARD-naive patients and patients with inadequate response to tumor necrosis factor inhibitors (TNFi-IR) with PsA who received bimekizumab (160 mg every 4 weeks; 431 and 267 patients, respectively) and guselkumab (100 mg every 4 weeks or every 8 weeks; 495 and 189 patients, respectively). They demonstrate that in biological DMARD-naive patients, bimekizumab was associated with a greater likelihood of achieving ≥70% improvement in American College of Rheumatology (ACR) response and minimal disease activity outcome at week 52 compared with guselkumab. Similar outcomes were observed in the TNFi-IR subgroup. Thus, bimekizumab may be more effective than guselkumab in PsA. Formal head-to-head studies comparing bimekizumab vs guselkumab are required.

 

With the availability of multiple targeted therapies for PsA, choosing the most effective and safe drug for a patient is difficult, especially in the absence of many head-to-head clinical trials. To help address this problem, Lin and Ren conducted a network meta-analysis of head-to-head active comparison studies in PsA. They included 17 studies in their analysis and demonstrated that Janus kinase inhibitors had the highest probability of achieving ACR 20/50/70 response. Treatment with IL-17A inhibitors was more likely than TNFi therapy to lead to resolution of enthesitis and dactylitis and achieving combined ACR 50 and Psoriasis Area Severity Index 100 response. Patients receiving phosphodiesterase 4 inhibitors were least likely to have adverse events. They conclude that when both efficacy and safety are considered, IL-17A inhibitors may be the better agent for initial therapy for PsA. IL-17A inhibitors are indeed safe and efficacious in PsA; more direct head-to-head comparisons as well as strategy trials are required to determine choice of first and subsequent therapy in PsA.

 

Infections are the most important adverse effects of targeted therapies. The risk for infection in PsA in real-world settings is not well known. In a cohort study that included 12,071 patients with PsA from the French national health insurance database who were new users of targeted therapies (adalimumab, etanercept, golimumab, certolizumab pegol, infliximab, secukinumab, ixekizumab, ustekinumab, and tofacitinib), Bastard and colleagues demonstrated that the incidence of serious infections in users of targeted therapies was 17.0 per 1000 person-years. Compared with new users of adalimumab, the risk for serious infections was significantly lower in new users of etanercept (weighted hazard ratio [wHR] 0.72; 95% CI 0.53-0.97) and ustekinumab (wHR 0.57; 95% CI 0.35-0.93). Thus, the overall risk for serious infections is low, with etanercept and ustekinumab being safer treatment options than adalimumab.

Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Publications
Topics
Sections
Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Dr. Chandran scans the journals, so you don't have to!
Dr. Chandran scans the journals, so you don't have to!

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Papers on psoriatic arthritis (PsA) published this month have focused on the clinical characteristics of PsA and pharmacologic treatment. Persistent inflammation leads to joint damage that is initially evident on imaging. Hen and colleagues evaluated 122 newly diagnosed, disease-modifying antirheumatic drug (DMARD)–naive patients with early PsA from the Leeds Spondyloarthropathy Register for Research and Observation cohort using conventional radiography and ultrasonography. Overall, 4655 hand and feet joints were assessed in 122 patients, of whom 24.6% had bone erosions at baseline; higher disease activity was observed in patients who did vs those who did not have bone erosions (P < .05). The prevalence of erosions was less in patients who had PsA symptoms < 8 months vs > 24 months (17.5% vs 24.3%, respectively). The agreement between conventional radiography and ultrasonography was high, with conventional radiography detecting more erosions. Thus, joint damage is seen early in patients with PsA; making a diagnosis within 8 months of symptoms is likely to lead to less joint damage and better outcomes.

 

Bimekizumab is a novel biologic therapy that inhibits interleukin (IL)–17A and IL-17F and is efficacious in the treatment of psoriasis, PsA, and axial spondyloarthritis. In the absence of a formal head-to-head study, matching-adjusted indirect comparison is a method to evaluate comparative effectiveness. Warren and colleagues ran a study that included biological DMARD-naive patients and patients with inadequate response to tumor necrosis factor inhibitors (TNFi-IR) with PsA who received bimekizumab (160 mg every 4 weeks; 431 and 267 patients, respectively) and guselkumab (100 mg every 4 weeks or every 8 weeks; 495 and 189 patients, respectively). They demonstrate that in biological DMARD-naive patients, bimekizumab was associated with a greater likelihood of achieving ≥70% improvement in American College of Rheumatology (ACR) response and minimal disease activity outcome at week 52 compared with guselkumab. Similar outcomes were observed in the TNFi-IR subgroup. Thus, bimekizumab may be more effective than guselkumab in PsA. Formal head-to-head studies comparing bimekizumab vs guselkumab are required.

 

With the availability of multiple targeted therapies for PsA, choosing the most effective and safe drug for a patient is difficult, especially in the absence of many head-to-head clinical trials. To help address this problem, Lin and Ren conducted a network meta-analysis of head-to-head active comparison studies in PsA. They included 17 studies in their analysis and demonstrated that Janus kinase inhibitors had the highest probability of achieving ACR 20/50/70 response. Treatment with IL-17A inhibitors was more likely than TNFi therapy to lead to resolution of enthesitis and dactylitis and achieving combined ACR 50 and Psoriasis Area Severity Index 100 response. Patients receiving phosphodiesterase 4 inhibitors were least likely to have adverse events. They conclude that when both efficacy and safety are considered, IL-17A inhibitors may be the better agent for initial therapy for PsA. IL-17A inhibitors are indeed safe and efficacious in PsA; more direct head-to-head comparisons as well as strategy trials are required to determine choice of first and subsequent therapy in PsA.

 

Infections are the most important adverse effects of targeted therapies. The risk for infection in PsA in real-world settings is not well known. In a cohort study that included 12,071 patients with PsA from the French national health insurance database who were new users of targeted therapies (adalimumab, etanercept, golimumab, certolizumab pegol, infliximab, secukinumab, ixekizumab, ustekinumab, and tofacitinib), Bastard and colleagues demonstrated that the incidence of serious infections in users of targeted therapies was 17.0 per 1000 person-years. Compared with new users of adalimumab, the risk for serious infections was significantly lower in new users of etanercept (weighted hazard ratio [wHR] 0.72; 95% CI 0.53-0.97) and ustekinumab (wHR 0.57; 95% CI 0.35-0.93). Thus, the overall risk for serious infections is low, with etanercept and ustekinumab being safer treatment options than adalimumab.

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Papers on psoriatic arthritis (PsA) published this month have focused on the clinical characteristics of PsA and pharmacologic treatment. Persistent inflammation leads to joint damage that is initially evident on imaging. Hen and colleagues evaluated 122 newly diagnosed, disease-modifying antirheumatic drug (DMARD)–naive patients with early PsA from the Leeds Spondyloarthropathy Register for Research and Observation cohort using conventional radiography and ultrasonography. Overall, 4655 hand and feet joints were assessed in 122 patients, of whom 24.6% had bone erosions at baseline; higher disease activity was observed in patients who did vs those who did not have bone erosions (P < .05). The prevalence of erosions was less in patients who had PsA symptoms < 8 months vs > 24 months (17.5% vs 24.3%, respectively). The agreement between conventional radiography and ultrasonography was high, with conventional radiography detecting more erosions. Thus, joint damage is seen early in patients with PsA; making a diagnosis within 8 months of symptoms is likely to lead to less joint damage and better outcomes.

 

Bimekizumab is a novel biologic therapy that inhibits interleukin (IL)–17A and IL-17F and is efficacious in the treatment of psoriasis, PsA, and axial spondyloarthritis. In the absence of a formal head-to-head study, matching-adjusted indirect comparison is a method to evaluate comparative effectiveness. Warren and colleagues ran a study that included biological DMARD-naive patients and patients with inadequate response to tumor necrosis factor inhibitors (TNFi-IR) with PsA who received bimekizumab (160 mg every 4 weeks; 431 and 267 patients, respectively) and guselkumab (100 mg every 4 weeks or every 8 weeks; 495 and 189 patients, respectively). They demonstrate that in biological DMARD-naive patients, bimekizumab was associated with a greater likelihood of achieving ≥70% improvement in American College of Rheumatology (ACR) response and minimal disease activity outcome at week 52 compared with guselkumab. Similar outcomes were observed in the TNFi-IR subgroup. Thus, bimekizumab may be more effective than guselkumab in PsA. Formal head-to-head studies comparing bimekizumab vs guselkumab are required.

 

With the availability of multiple targeted therapies for PsA, choosing the most effective and safe drug for a patient is difficult, especially in the absence of many head-to-head clinical trials. To help address this problem, Lin and Ren conducted a network meta-analysis of head-to-head active comparison studies in PsA. They included 17 studies in their analysis and demonstrated that Janus kinase inhibitors had the highest probability of achieving ACR 20/50/70 response. Treatment with IL-17A inhibitors was more likely than TNFi therapy to lead to resolution of enthesitis and dactylitis and achieving combined ACR 50 and Psoriasis Area Severity Index 100 response. Patients receiving phosphodiesterase 4 inhibitors were least likely to have adverse events. They conclude that when both efficacy and safety are considered, IL-17A inhibitors may be the better agent for initial therapy for PsA. IL-17A inhibitors are indeed safe and efficacious in PsA; more direct head-to-head comparisons as well as strategy trials are required to determine choice of first and subsequent therapy in PsA.

 

Infections are the most important adverse effects of targeted therapies. The risk for infection in PsA in real-world settings is not well known. In a cohort study that included 12,071 patients with PsA from the French national health insurance database who were new users of targeted therapies (adalimumab, etanercept, golimumab, certolizumab pegol, infliximab, secukinumab, ixekizumab, ustekinumab, and tofacitinib), Bastard and colleagues demonstrated that the incidence of serious infections in users of targeted therapies was 17.0 per 1000 person-years. Compared with new users of adalimumab, the risk for serious infections was significantly lower in new users of etanercept (weighted hazard ratio [wHR] 0.72; 95% CI 0.53-0.97) and ustekinumab (wHR 0.57; 95% CI 0.35-0.93). Thus, the overall risk for serious infections is low, with etanercept and ustekinumab being safer treatment options than adalimumab.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 04/05/2021 - 09:15
Un-Gate On Date
Mon, 04/05/2021 - 09:15
Use ProPublica
CFC Schedule Remove Status
Mon, 04/05/2021 - 09:15
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

Commentary: Studies Often Do Not Answer Clinical Questions in AD, May 2024

Article Type
Changed
Fri, 04/26/2024 - 15:45
Dr. Feldman scans the journals, so you don’t have to!

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E

In "Atopic Dermatitis in Early Childhood and Risk of Inflammatory Bowel Disease: A Scandinavian Birth Cohort Study," Lerchova and colleagues found a statistically significant increased risk for inflammatory bowel disease (IBD) in children with atopic dermatitis. The study had a large patient population, giving it the power to identify very small differences. The researchers found increased risks for IBD, Crohn's disease, and ulcerative colitis (UC) in children with atopic dermatitis; UC had the greatest relative risk. But I don't think this risk was clinically meaningful. About 2 in every 1000 children with atopic dermatitis had UC, whereas about 1 in every 1000 children without atopic dermatitis had UC. Even if the increased absolute risk of 1 in 1000 children was due to atopic dermatitis and not to other factors, I don't think it justifies the authors' conclusion that "these findings might be useful in identifying at-risk individuals for IBD."

Sometimes reviewing articles makes me feel like a crotchety old man. A study by Guttman-Yassky and colleagues, "Targeting IL-13 With Tralokinumab Normalizes Type 2 Inflammation in Atopic Dermatitis Both Early and at 2 Years," didn't seem to test any specific hypothesis. The researchers just looked at a variety of inflammation markers in patients with atopic dermatitis treated with tralokinumab, an interleukin-13 (IL-13) antagonist. In these patients, as expected, the atopic dermatitis improved; so did the inflammatory markers. Did we learn anything clinically useful? I don't think so. We already know that IL-13 is important in atopic dermatitis because when we block IL-13, atopic dermatitis improves.


Vitamin D supplementation doesn't appear to improve atopic dermatitis, as reported by Borzutzky and colleagues in "Effect of Weekly Vitamin D Supplementation on the Severity of Atopic Dermatitis and Type 2 Immunity Biomarkers in Children: A Randomized Controlled Trial." A group of 101 children with atopic dermatitis were randomly assigned to receive oral vitamin D supplementation or placebo. The two groups improved to a similar extent. If you know me, you know I'm wondering whether they took the medication. It appears that they did, because at baseline most of the children were vitamin D deficient, and vitamin D levels improved greatly in the group treated with vitamin D but not in the placebo group.


Journals such as the Journal of the American Academy of Dermatology should require articles to report absolute risk. In "Risk of Lymphoma in Patients With Atopic Dermatitis: A Case-Control Study in the All of Us Database," Powers and colleagues tell us that atopic dermatitis is associated with a statistically significantly increased risk for lymphoma. This means that increased risk wasn't likely due to chance alone. The article says nothing, as far as I could tell, about how big the risk is. Does everyone get lymphoma? Or is it a one in a million risk? Without knowing the absolute risk, the relative risk doesn't tell us whether there is a clinically meaningful increased risk or not. I suspect the increased risk is small. If the incidence of lymphoma is about 2 in 10,000 and peripheral T-cell lymphomas (PTCL) account for 10% of those, even a fourfold increase in the risk for PTCL (the form of lymphoma with the highest relative risk) would not amount to much. 


Traidl and colleagues report in "Treatment of Moderate-to-Severe Atopic Dermatitis With Baricitinib: Results From an Interim Analysis of the TREATgermany Registry" that the Janus kinase inhibitor baricitinib makes atopic dermatitis better. 


In "Dupilumab Therapy for Atopic Dermatitis Is Associated With Increased Risk of Cutaneous T Cell Lymphoma," Hasan and colleagues report that "it requires 738 prescriptions of dupilumab to produce one case of CTCL [cutaneous T-cell lymphoma]." It seems that this finding could easily be due to 1 in 738 people with a rash thought to be severe atopic dermatitis needing dupilumab having CTCL, not atopic dermatitis, to begin with. If we were to wonder whether dupilumab causes CTCL, perhaps it would be better to study asthma patients treated with or without dupilumab.
 

Author and Disclosure Information

Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

He has reported no disclosures.

Publications
Topics
Sections
Author and Disclosure Information

Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

He has reported no disclosures.

Author and Disclosure Information

Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

He has reported no disclosures.

Dr. Feldman scans the journals, so you don’t have to!
Dr. Feldman scans the journals, so you don’t have to!

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E

In "Atopic Dermatitis in Early Childhood and Risk of Inflammatory Bowel Disease: A Scandinavian Birth Cohort Study," Lerchova and colleagues found a statistically significant increased risk for inflammatory bowel disease (IBD) in children with atopic dermatitis. The study had a large patient population, giving it the power to identify very small differences. The researchers found increased risks for IBD, Crohn's disease, and ulcerative colitis (UC) in children with atopic dermatitis; UC had the greatest relative risk. But I don't think this risk was clinically meaningful. About 2 in every 1000 children with atopic dermatitis had UC, whereas about 1 in every 1000 children without atopic dermatitis had UC. Even if the increased absolute risk of 1 in 1000 children was due to atopic dermatitis and not to other factors, I don't think it justifies the authors' conclusion that "these findings might be useful in identifying at-risk individuals for IBD."

Sometimes reviewing articles makes me feel like a crotchety old man. A study by Guttman-Yassky and colleagues, "Targeting IL-13 With Tralokinumab Normalizes Type 2 Inflammation in Atopic Dermatitis Both Early and at 2 Years," didn't seem to test any specific hypothesis. The researchers just looked at a variety of inflammation markers in patients with atopic dermatitis treated with tralokinumab, an interleukin-13 (IL-13) antagonist. In these patients, as expected, the atopic dermatitis improved; so did the inflammatory markers. Did we learn anything clinically useful? I don't think so. We already know that IL-13 is important in atopic dermatitis because when we block IL-13, atopic dermatitis improves.


Vitamin D supplementation doesn't appear to improve atopic dermatitis, as reported by Borzutzky and colleagues in "Effect of Weekly Vitamin D Supplementation on the Severity of Atopic Dermatitis and Type 2 Immunity Biomarkers in Children: A Randomized Controlled Trial." A group of 101 children with atopic dermatitis were randomly assigned to receive oral vitamin D supplementation or placebo. The two groups improved to a similar extent. If you know me, you know I'm wondering whether they took the medication. It appears that they did, because at baseline most of the children were vitamin D deficient, and vitamin D levels improved greatly in the group treated with vitamin D but not in the placebo group.


Journals such as the Journal of the American Academy of Dermatology should require articles to report absolute risk. In "Risk of Lymphoma in Patients With Atopic Dermatitis: A Case-Control Study in the All of Us Database," Powers and colleagues tell us that atopic dermatitis is associated with a statistically significantly increased risk for lymphoma. This means that increased risk wasn't likely due to chance alone. The article says nothing, as far as I could tell, about how big the risk is. Does everyone get lymphoma? Or is it a one in a million risk? Without knowing the absolute risk, the relative risk doesn't tell us whether there is a clinically meaningful increased risk or not. I suspect the increased risk is small. If the incidence of lymphoma is about 2 in 10,000 and peripheral T-cell lymphomas (PTCL) account for 10% of those, even a fourfold increase in the risk for PTCL (the form of lymphoma with the highest relative risk) would not amount to much. 


Traidl and colleagues report in "Treatment of Moderate-to-Severe Atopic Dermatitis With Baricitinib: Results From an Interim Analysis of the TREATgermany Registry" that the Janus kinase inhibitor baricitinib makes atopic dermatitis better. 


In "Dupilumab Therapy for Atopic Dermatitis Is Associated With Increased Risk of Cutaneous T Cell Lymphoma," Hasan and colleagues report that "it requires 738 prescriptions of dupilumab to produce one case of CTCL [cutaneous T-cell lymphoma]." It seems that this finding could easily be due to 1 in 738 people with a rash thought to be severe atopic dermatitis needing dupilumab having CTCL, not atopic dermatitis, to begin with. If we were to wonder whether dupilumab causes CTCL, perhaps it would be better to study asthma patients treated with or without dupilumab.
 

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E

In "Atopic Dermatitis in Early Childhood and Risk of Inflammatory Bowel Disease: A Scandinavian Birth Cohort Study," Lerchova and colleagues found a statistically significant increased risk for inflammatory bowel disease (IBD) in children with atopic dermatitis. The study had a large patient population, giving it the power to identify very small differences. The researchers found increased risks for IBD, Crohn's disease, and ulcerative colitis (UC) in children with atopic dermatitis; UC had the greatest relative risk. But I don't think this risk was clinically meaningful. About 2 in every 1000 children with atopic dermatitis had UC, whereas about 1 in every 1000 children without atopic dermatitis had UC. Even if the increased absolute risk of 1 in 1000 children was due to atopic dermatitis and not to other factors, I don't think it justifies the authors' conclusion that "these findings might be useful in identifying at-risk individuals for IBD."

Sometimes reviewing articles makes me feel like a crotchety old man. A study by Guttman-Yassky and colleagues, "Targeting IL-13 With Tralokinumab Normalizes Type 2 Inflammation in Atopic Dermatitis Both Early and at 2 Years," didn't seem to test any specific hypothesis. The researchers just looked at a variety of inflammation markers in patients with atopic dermatitis treated with tralokinumab, an interleukin-13 (IL-13) antagonist. In these patients, as expected, the atopic dermatitis improved; so did the inflammatory markers. Did we learn anything clinically useful? I don't think so. We already know that IL-13 is important in atopic dermatitis because when we block IL-13, atopic dermatitis improves.


Vitamin D supplementation doesn't appear to improve atopic dermatitis, as reported by Borzutzky and colleagues in "Effect of Weekly Vitamin D Supplementation on the Severity of Atopic Dermatitis and Type 2 Immunity Biomarkers in Children: A Randomized Controlled Trial." A group of 101 children with atopic dermatitis were randomly assigned to receive oral vitamin D supplementation or placebo. The two groups improved to a similar extent. If you know me, you know I'm wondering whether they took the medication. It appears that they did, because at baseline most of the children were vitamin D deficient, and vitamin D levels improved greatly in the group treated with vitamin D but not in the placebo group.


Journals such as the Journal of the American Academy of Dermatology should require articles to report absolute risk. In "Risk of Lymphoma in Patients With Atopic Dermatitis: A Case-Control Study in the All of Us Database," Powers and colleagues tell us that atopic dermatitis is associated with a statistically significantly increased risk for lymphoma. This means that increased risk wasn't likely due to chance alone. The article says nothing, as far as I could tell, about how big the risk is. Does everyone get lymphoma? Or is it a one in a million risk? Without knowing the absolute risk, the relative risk doesn't tell us whether there is a clinically meaningful increased risk or not. I suspect the increased risk is small. If the incidence of lymphoma is about 2 in 10,000 and peripheral T-cell lymphomas (PTCL) account for 10% of those, even a fourfold increase in the risk for PTCL (the form of lymphoma with the highest relative risk) would not amount to much. 


Traidl and colleagues report in "Treatment of Moderate-to-Severe Atopic Dermatitis With Baricitinib: Results From an Interim Analysis of the TREATgermany Registry" that the Janus kinase inhibitor baricitinib makes atopic dermatitis better. 


In "Dupilumab Therapy for Atopic Dermatitis Is Associated With Increased Risk of Cutaneous T Cell Lymphoma," Hasan and colleagues report that "it requires 738 prescriptions of dupilumab to produce one case of CTCL [cutaneous T-cell lymphoma]." It seems that this finding could easily be due to 1 in 738 people with a rash thought to be severe atopic dermatitis needing dupilumab having CTCL, not atopic dermatitis, to begin with. If we were to wonder whether dupilumab causes CTCL, perhaps it would be better to study asthma patients treated with or without dupilumab.
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 03/26/2024 - 15:00
Un-Gate On Date
Tue, 03/26/2024 - 15:00
Use ProPublica
CFC Schedule Remove Status
Tue, 03/26/2024 - 15:00
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

Commentary: Gut Dysbiosis, DMARD, Joint Involvement, and MACE in PsA, April 2024

Article Type
Changed
Mon, 04/01/2024 - 12:37
Dr. Chandran scans the journals, so you don't have to!

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20lang%3D%22EN-CA%22%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Prior studies have demonstrated an association between gut dysbiosis and psoriatic arthritis (PsA). It is difficult, however, to determine causal associations by cross-sectional studies. Mendelian randomization is an approach that uses genetic variants to assess causal relationships using observational data. Xu and colleagues used this approach to analyze summary-level data of gut microbiota taxa (n = 18,340), PsA (n = 339,050), and metabolites (n = 7824) from participants included in the MiBioGen consortium, FinnGen Biobank, and TwinsUK and KORA cohorts, respectively. Adjusted multivariable Mendelian randomization analysis showed that a higher relative abundance of microbiota belonging to the family Rikenellaceae (odds ratio [OR] 0.5; 95% CI 0.320-0.780) and elevated serum levels of X-11538 (OR 0.448; 95% CI 0.244-0.821) were causally associated with a reduced risk for PsA. The study highlights the potential role of gut microbiota in PsA susceptibility and a possible means for primary prevention of PsA.

 

After PsA onset, early diagnosis and management leads to better long-term outcomes. These prior observations were confirmed in a study by Snoeck Henkemans and colleagues that included 708 newly diagnosed patients with PsA naive to disease-modifying antirheumatic drugs (DMARD) who were followed up for 3 years or more. Patients with a short (<12 weeks) vs long delay (>1 year) in PsA diagnosis after symptom onset were more likely to achieve minimum disease activity (OR 2.55; 95% CI 1.37-4.76). Thus, longer delay in diagnosing PsA is associated with worse clinical outcomes.

 

Bimekizumab is a novel biologic therapy that inhibits interleukins (IL)-17A and -17F and is efficacious in the treatment of psoriasis, PsA, and axial spondyloarthritis. However, the effectiveness in PsA vis-à-vis other IL-17A inhibitors is not known. In the absence of a formal head-to-head study, matching-adjusted indirect comparisons is a method to evaluate comparative effectiveness. Such a study by Mease and colleagues included the data of patients with PsA who were biological DMARD–naive or who had an inadequate response to tumor necrosis factor inhibitors (TNFi-IR), and who received bimekizumab from the BE OPTIMAL (n = 236) and BE COMPLETE (n = 146) trials and secukinumab from the FUTURE 2 trial (n = 200). They demonstrated that, in the biological DMARD–naive subgroup, the probability of achieving at least 70% improvement in American College of Rheumatology (ACR) response was two times higher with bimekizumab (160 mg every 4 weeks) vs secukinumab (150 mg or 300 mg every 4 weeks) at week 52. In the TNFi-IR subgroup, bimekizumab had a greater likelihood of response compared with 150 mg secukinumab for ACR20, ACR70, and minimal disease activity outcomes and a greater likelihood of response compared with 300 mg secukinumab for ACR50 and minimal disease activity. Thus, bimekizumab is at least as effective as secukinumab in PsA. Formal head-to-head studies comparing bimekizumab with other IL-17A inhibitors are required.

 

Distal interphalangeal (DIP) joint involvement is an important manifestation of PsA and is closely related to nail dystrophy in the adjacent nail. Ixekizumab is another biologic that targets IL-17A. In a post hoc analysis of the SPIRIT-H2H study, McGonagle and colleagues confirmed that over 96% of patients with PsA and simultaneous DIP joint involvement reported adjacent nail psoriasis. When compared with adalimumab, ixekizumab led to greater improvements in DIP involvement and adjacent nail psoriasis as early as week 12 (38.8% vs 28.4%; P < .0001), with improvements sustained up to week 52 (64.9% vs 57.5%; P = .0055). This probably reflects a greater effectiveness of IL-17A inhibition in treating skin and nail psoriasis compared with TNFi.

 

Finally, in a population-based retrospective cohort study that included 13,905 patients with PsA (n = 1672) or rheumatoid arthritis (n = 12,233) who did not have any previous history of major adverse cardiovascular events (MACE), Meng and colleagues showed that the incidence rates of MACE were similar in patients with PsA and rheumatoid arthritis. Thus, cardiovascular risk management should be similarly aggressive in patients with PsA and rheumatoid arthritis.

Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Publications
Topics
Sections
Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Dr. Chandran scans the journals, so you don't have to!
Dr. Chandran scans the journals, so you don't have to!

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20lang%3D%22EN-CA%22%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Prior studies have demonstrated an association between gut dysbiosis and psoriatic arthritis (PsA). It is difficult, however, to determine causal associations by cross-sectional studies. Mendelian randomization is an approach that uses genetic variants to assess causal relationships using observational data. Xu and colleagues used this approach to analyze summary-level data of gut microbiota taxa (n = 18,340), PsA (n = 339,050), and metabolites (n = 7824) from participants included in the MiBioGen consortium, FinnGen Biobank, and TwinsUK and KORA cohorts, respectively. Adjusted multivariable Mendelian randomization analysis showed that a higher relative abundance of microbiota belonging to the family Rikenellaceae (odds ratio [OR] 0.5; 95% CI 0.320-0.780) and elevated serum levels of X-11538 (OR 0.448; 95% CI 0.244-0.821) were causally associated with a reduced risk for PsA. The study highlights the potential role of gut microbiota in PsA susceptibility and a possible means for primary prevention of PsA.

 

After PsA onset, early diagnosis and management leads to better long-term outcomes. These prior observations were confirmed in a study by Snoeck Henkemans and colleagues that included 708 newly diagnosed patients with PsA naive to disease-modifying antirheumatic drugs (DMARD) who were followed up for 3 years or more. Patients with a short (<12 weeks) vs long delay (>1 year) in PsA diagnosis after symptom onset were more likely to achieve minimum disease activity (OR 2.55; 95% CI 1.37-4.76). Thus, longer delay in diagnosing PsA is associated with worse clinical outcomes.

 

Bimekizumab is a novel biologic therapy that inhibits interleukins (IL)-17A and -17F and is efficacious in the treatment of psoriasis, PsA, and axial spondyloarthritis. However, the effectiveness in PsA vis-à-vis other IL-17A inhibitors is not known. In the absence of a formal head-to-head study, matching-adjusted indirect comparisons is a method to evaluate comparative effectiveness. Such a study by Mease and colleagues included the data of patients with PsA who were biological DMARD–naive or who had an inadequate response to tumor necrosis factor inhibitors (TNFi-IR), and who received bimekizumab from the BE OPTIMAL (n = 236) and BE COMPLETE (n = 146) trials and secukinumab from the FUTURE 2 trial (n = 200). They demonstrated that, in the biological DMARD–naive subgroup, the probability of achieving at least 70% improvement in American College of Rheumatology (ACR) response was two times higher with bimekizumab (160 mg every 4 weeks) vs secukinumab (150 mg or 300 mg every 4 weeks) at week 52. In the TNFi-IR subgroup, bimekizumab had a greater likelihood of response compared with 150 mg secukinumab for ACR20, ACR70, and minimal disease activity outcomes and a greater likelihood of response compared with 300 mg secukinumab for ACR50 and minimal disease activity. Thus, bimekizumab is at least as effective as secukinumab in PsA. Formal head-to-head studies comparing bimekizumab with other IL-17A inhibitors are required.

 

Distal interphalangeal (DIP) joint involvement is an important manifestation of PsA and is closely related to nail dystrophy in the adjacent nail. Ixekizumab is another biologic that targets IL-17A. In a post hoc analysis of the SPIRIT-H2H study, McGonagle and colleagues confirmed that over 96% of patients with PsA and simultaneous DIP joint involvement reported adjacent nail psoriasis. When compared with adalimumab, ixekizumab led to greater improvements in DIP involvement and adjacent nail psoriasis as early as week 12 (38.8% vs 28.4%; P < .0001), with improvements sustained up to week 52 (64.9% vs 57.5%; P = .0055). This probably reflects a greater effectiveness of IL-17A inhibition in treating skin and nail psoriasis compared with TNFi.

 

Finally, in a population-based retrospective cohort study that included 13,905 patients with PsA (n = 1672) or rheumatoid arthritis (n = 12,233) who did not have any previous history of major adverse cardiovascular events (MACE), Meng and colleagues showed that the incidence rates of MACE were similar in patients with PsA and rheumatoid arthritis. Thus, cardiovascular risk management should be similarly aggressive in patients with PsA and rheumatoid arthritis.

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20lang%3D%22EN-CA%22%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Prior studies have demonstrated an association between gut dysbiosis and psoriatic arthritis (PsA). It is difficult, however, to determine causal associations by cross-sectional studies. Mendelian randomization is an approach that uses genetic variants to assess causal relationships using observational data. Xu and colleagues used this approach to analyze summary-level data of gut microbiota taxa (n = 18,340), PsA (n = 339,050), and metabolites (n = 7824) from participants included in the MiBioGen consortium, FinnGen Biobank, and TwinsUK and KORA cohorts, respectively. Adjusted multivariable Mendelian randomization analysis showed that a higher relative abundance of microbiota belonging to the family Rikenellaceae (odds ratio [OR] 0.5; 95% CI 0.320-0.780) and elevated serum levels of X-11538 (OR 0.448; 95% CI 0.244-0.821) were causally associated with a reduced risk for PsA. The study highlights the potential role of gut microbiota in PsA susceptibility and a possible means for primary prevention of PsA.

 

After PsA onset, early diagnosis and management leads to better long-term outcomes. These prior observations were confirmed in a study by Snoeck Henkemans and colleagues that included 708 newly diagnosed patients with PsA naive to disease-modifying antirheumatic drugs (DMARD) who were followed up for 3 years or more. Patients with a short (<12 weeks) vs long delay (>1 year) in PsA diagnosis after symptom onset were more likely to achieve minimum disease activity (OR 2.55; 95% CI 1.37-4.76). Thus, longer delay in diagnosing PsA is associated with worse clinical outcomes.

 

Bimekizumab is a novel biologic therapy that inhibits interleukins (IL)-17A and -17F and is efficacious in the treatment of psoriasis, PsA, and axial spondyloarthritis. However, the effectiveness in PsA vis-à-vis other IL-17A inhibitors is not known. In the absence of a formal head-to-head study, matching-adjusted indirect comparisons is a method to evaluate comparative effectiveness. Such a study by Mease and colleagues included the data of patients with PsA who were biological DMARD–naive or who had an inadequate response to tumor necrosis factor inhibitors (TNFi-IR), and who received bimekizumab from the BE OPTIMAL (n = 236) and BE COMPLETE (n = 146) trials and secukinumab from the FUTURE 2 trial (n = 200). They demonstrated that, in the biological DMARD–naive subgroup, the probability of achieving at least 70% improvement in American College of Rheumatology (ACR) response was two times higher with bimekizumab (160 mg every 4 weeks) vs secukinumab (150 mg or 300 mg every 4 weeks) at week 52. In the TNFi-IR subgroup, bimekizumab had a greater likelihood of response compared with 150 mg secukinumab for ACR20, ACR70, and minimal disease activity outcomes and a greater likelihood of response compared with 300 mg secukinumab for ACR50 and minimal disease activity. Thus, bimekizumab is at least as effective as secukinumab in PsA. Formal head-to-head studies comparing bimekizumab with other IL-17A inhibitors are required.

 

Distal interphalangeal (DIP) joint involvement is an important manifestation of PsA and is closely related to nail dystrophy in the adjacent nail. Ixekizumab is another biologic that targets IL-17A. In a post hoc analysis of the SPIRIT-H2H study, McGonagle and colleagues confirmed that over 96% of patients with PsA and simultaneous DIP joint involvement reported adjacent nail psoriasis. When compared with adalimumab, ixekizumab led to greater improvements in DIP involvement and adjacent nail psoriasis as early as week 12 (38.8% vs 28.4%; P < .0001), with improvements sustained up to week 52 (64.9% vs 57.5%; P = .0055). This probably reflects a greater effectiveness of IL-17A inhibition in treating skin and nail psoriasis compared with TNFi.

 

Finally, in a population-based retrospective cohort study that included 13,905 patients with PsA (n = 1672) or rheumatoid arthritis (n = 12,233) who did not have any previous history of major adverse cardiovascular events (MACE), Meng and colleagues showed that the incidence rates of MACE were similar in patients with PsA and rheumatoid arthritis. Thus, cardiovascular risk management should be similarly aggressive in patients with PsA and rheumatoid arthritis.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 04/05/2021 - 09:15
Un-Gate On Date
Mon, 04/05/2021 - 09:15
Use ProPublica
CFC Schedule Remove Status
Mon, 04/05/2021 - 09:15
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
Activity Salesforce Deliverable ID
400312.1
Activity ID
110008
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
SKYRIZI [ 5052 ]

Commentary: MRI Surveillance and Risk Factors in Breast Cancer, April 2024

Article Type
Changed
Mon, 04/29/2024 - 18:04
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Women with pathogenic BRCA1/2 mutations are presented options of risk-reducing surgery or enhanced surveillance to address their elevated lifetime risk for breast cancer. In regard to breast cancer screening for these women, guidelines recommend annual mammography and breast MRI for those aged 30-75 years; for younger women (age 25-29 years), annual MRI or an individualized schedule on the basis of family history if a breast cancer diagnosis before age 30 is present.[1] Prior studies have highlighted the role of screening MRI in "downstaging," meaning MRI screening detected breast cancers at an earlier stage vs those identified with mammography.[2] As with any screening tool, it is essential to demonstrate the effect of MRI surveillance on mortality for women with BRCA mutations. A cohort study that included 2488 women (age ≥ 30 years) with a BRCA1 (n = 2004) or BRCA2 (n = 484) mutation compared breast cancer mortality rates among those women who participated in MRI screening with those who did not (Lubinski et al). After a median follow-up of 9.2 years, 344 women (13.8%) developed breast cancer, and 35 (1.4%) died from breast cancer. There was an 80% reduction in breast cancer mortality among BRCA1 mutation carriers who participated in MRI surveillance vs those who did not (age-adjusted hazard ratio [HR] 0.20; 95% CI 0.10-0.43; P < .001), but this was not observed for women with BRCA2 mutations (age-adjusted HR 0.87; 95% CI 0.10-17.25; P = .93). At 20 years, the breast cancer mortality rate was 3.2% in the MRI surveillance group compared with 14.9% in the group who did not undergo surveillance. A separate cohort study from Ontario, Canada, including 489 women with BRCA1/2 pathogenic mutations found a 2.0% rate of breast cancer-related mortality at 20 years after the first MRI screening.[3] These data support an intensified surveillance schedule for BRCA mutation carriers, with a need for further research and insight in the BRCA2 population.

A positive family history of cancer and obesity are established risk factors for development of breast cancer among women.[4,5] A population-based cohort study that included 15,055 Chinese women evaluated the association and interaction between body mass index (BMI) and family history of cancer on the risk for breast cancer (Cao et al). The incidence risk for breast cancer was highest in the group with obesity vs the group with normal weight (adjusted HR 2.09; 95% CI 1.42-3.07), and those with a family history of cancer also had an increased risk vs those without a family history of cancer (adjusted HR 1.63; 95% CI 1.22-2.49). Furthermore, women with a BMI ≥ 24 and family history of cancer had a higher risk for breast cancer development compared with women with a BMI < 24 and no family history of cancer (adjusted HR 2.06; 95% CI 1.39-3.06). This study indicates a heightened breast cancer risk when cancer family history and obesity coexist, suggesting the importance of addressing modifiable risk factors and targeting lifestyle interventions in this population.

Triple-negative breast cancer (TNBC), although exhibiting its own heterogeneity, has various features that differentiate this subtype from luminal breast cancers. For example, TNBC generally has a more aggressive course, increased responsiveness to chemotherapy, and earlier pattern of recurrence compared with hormone receptor–positive disease. Prior studies have also shown that established breast cancer risk factors reflect those for the luminal A subtype, whereas those for TNBC are less consistent.[6] A meta-analysis that included 33 studies evaluated the association between traditional breast cancer risk factors and TNBC incidence (Kumar et al). Family history (odds ratio [OR] 1.55; 95% CI 1.34-1.81; P < .001), longer duration of oral contraceptive use (OR 1.29; 95% CI 1.08-1.55; P < .001), and higher breast density (OR 2.19; 95% CI 1.67-2.88; P < .001) were significantly associated with an increased risk for TNBC. Factors including later age at menarche, later age at first birth, and breastfeeding were associated with reduced risk for TNBC. Furthermore, there was no significant association with parity, menopausal hormone therapy, alcohol, smoking, and BMI. This study highlights distinct risk factors that may contribute to a higher risk for TNBC, and future research will be valuable to better elucidate the mechanisms at play and to further understand the differences within this subtype itself.

Additional References

  1. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic. Version 3.2024. Source 
  2. Saadatmand S, Geuzinge HA, Rutgers EJT, et al; on behalf of the FaMRIsc study group. MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): A multicentre, randomised, controlled trial. Lancet Oncol. 2019;20:1136-1147. doi: 10.1016/S1470-2045(19)30275-X  Source
  3. Warner E, Zhu S, Plewes DB, et al. Breast cancer mortality among women with a BRCA1 or BRCA2 mutation in a magnetic resonance imaging plus mammography screening program. Cancers (Basel). 2020;12:3479. doi: 10.3390/cancers12113479 Source
  4. Picon-Ruiz M, Morata-Tarifa C, Valle-Goffin JJ, et al. Obesity and adverse breast cancer risk and outcome: Mechanistic insights and strategies for intervention. CA Cancer J Clin. 2017;67:378-397. doi: 10.3322/caac.21405 Source
  5. Engmann NJ, Golmakani MK, Miglioretti DL, et al; for the Breast Cancer Surveillance Consortium. Population-attributable risk proportion of clinical risk factors for breast cancer. JAMA Oncol. 2017;3:1228-1236. doi: 10.1001/jamaoncol.2016.6326 Source
  6. Barnard ME, Boeke CE, Tamimi RM. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim Biophys Acta Rev Cancer. 2015;1856:73-85. doi: 10.1016/j.bbcan.2015.0002 Source
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Women with pathogenic BRCA1/2 mutations are presented options of risk-reducing surgery or enhanced surveillance to address their elevated lifetime risk for breast cancer. In regard to breast cancer screening for these women, guidelines recommend annual mammography and breast MRI for those aged 30-75 years; for younger women (age 25-29 years), annual MRI or an individualized schedule on the basis of family history if a breast cancer diagnosis before age 30 is present.[1] Prior studies have highlighted the role of screening MRI in "downstaging," meaning MRI screening detected breast cancers at an earlier stage vs those identified with mammography.[2] As with any screening tool, it is essential to demonstrate the effect of MRI surveillance on mortality for women with BRCA mutations. A cohort study that included 2488 women (age ≥ 30 years) with a BRCA1 (n = 2004) or BRCA2 (n = 484) mutation compared breast cancer mortality rates among those women who participated in MRI screening with those who did not (Lubinski et al). After a median follow-up of 9.2 years, 344 women (13.8%) developed breast cancer, and 35 (1.4%) died from breast cancer. There was an 80% reduction in breast cancer mortality among BRCA1 mutation carriers who participated in MRI surveillance vs those who did not (age-adjusted hazard ratio [HR] 0.20; 95% CI 0.10-0.43; P < .001), but this was not observed for women with BRCA2 mutations (age-adjusted HR 0.87; 95% CI 0.10-17.25; P = .93). At 20 years, the breast cancer mortality rate was 3.2% in the MRI surveillance group compared with 14.9% in the group who did not undergo surveillance. A separate cohort study from Ontario, Canada, including 489 women with BRCA1/2 pathogenic mutations found a 2.0% rate of breast cancer-related mortality at 20 years after the first MRI screening.[3] These data support an intensified surveillance schedule for BRCA mutation carriers, with a need for further research and insight in the BRCA2 population.

A positive family history of cancer and obesity are established risk factors for development of breast cancer among women.[4,5] A population-based cohort study that included 15,055 Chinese women evaluated the association and interaction between body mass index (BMI) and family history of cancer on the risk for breast cancer (Cao et al). The incidence risk for breast cancer was highest in the group with obesity vs the group with normal weight (adjusted HR 2.09; 95% CI 1.42-3.07), and those with a family history of cancer also had an increased risk vs those without a family history of cancer (adjusted HR 1.63; 95% CI 1.22-2.49). Furthermore, women with a BMI ≥ 24 and family history of cancer had a higher risk for breast cancer development compared with women with a BMI < 24 and no family history of cancer (adjusted HR 2.06; 95% CI 1.39-3.06). This study indicates a heightened breast cancer risk when cancer family history and obesity coexist, suggesting the importance of addressing modifiable risk factors and targeting lifestyle interventions in this population.

Triple-negative breast cancer (TNBC), although exhibiting its own heterogeneity, has various features that differentiate this subtype from luminal breast cancers. For example, TNBC generally has a more aggressive course, increased responsiveness to chemotherapy, and earlier pattern of recurrence compared with hormone receptor–positive disease. Prior studies have also shown that established breast cancer risk factors reflect those for the luminal A subtype, whereas those for TNBC are less consistent.[6] A meta-analysis that included 33 studies evaluated the association between traditional breast cancer risk factors and TNBC incidence (Kumar et al). Family history (odds ratio [OR] 1.55; 95% CI 1.34-1.81; P < .001), longer duration of oral contraceptive use (OR 1.29; 95% CI 1.08-1.55; P < .001), and higher breast density (OR 2.19; 95% CI 1.67-2.88; P < .001) were significantly associated with an increased risk for TNBC. Factors including later age at menarche, later age at first birth, and breastfeeding were associated with reduced risk for TNBC. Furthermore, there was no significant association with parity, menopausal hormone therapy, alcohol, smoking, and BMI. This study highlights distinct risk factors that may contribute to a higher risk for TNBC, and future research will be valuable to better elucidate the mechanisms at play and to further understand the differences within this subtype itself.

Additional References

  1. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic. Version 3.2024. Source 
  2. Saadatmand S, Geuzinge HA, Rutgers EJT, et al; on behalf of the FaMRIsc study group. MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): A multicentre, randomised, controlled trial. Lancet Oncol. 2019;20:1136-1147. doi: 10.1016/S1470-2045(19)30275-X  Source
  3. Warner E, Zhu S, Plewes DB, et al. Breast cancer mortality among women with a BRCA1 or BRCA2 mutation in a magnetic resonance imaging plus mammography screening program. Cancers (Basel). 2020;12:3479. doi: 10.3390/cancers12113479 Source
  4. Picon-Ruiz M, Morata-Tarifa C, Valle-Goffin JJ, et al. Obesity and adverse breast cancer risk and outcome: Mechanistic insights and strategies for intervention. CA Cancer J Clin. 2017;67:378-397. doi: 10.3322/caac.21405 Source
  5. Engmann NJ, Golmakani MK, Miglioretti DL, et al; for the Breast Cancer Surveillance Consortium. Population-attributable risk proportion of clinical risk factors for breast cancer. JAMA Oncol. 2017;3:1228-1236. doi: 10.1001/jamaoncol.2016.6326 Source
  6. Barnard ME, Boeke CE, Tamimi RM. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim Biophys Acta Rev Cancer. 2015;1856:73-85. doi: 10.1016/j.bbcan.2015.0002 Source

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Women with pathogenic BRCA1/2 mutations are presented options of risk-reducing surgery or enhanced surveillance to address their elevated lifetime risk for breast cancer. In regard to breast cancer screening for these women, guidelines recommend annual mammography and breast MRI for those aged 30-75 years; for younger women (age 25-29 years), annual MRI or an individualized schedule on the basis of family history if a breast cancer diagnosis before age 30 is present.[1] Prior studies have highlighted the role of screening MRI in "downstaging," meaning MRI screening detected breast cancers at an earlier stage vs those identified with mammography.[2] As with any screening tool, it is essential to demonstrate the effect of MRI surveillance on mortality for women with BRCA mutations. A cohort study that included 2488 women (age ≥ 30 years) with a BRCA1 (n = 2004) or BRCA2 (n = 484) mutation compared breast cancer mortality rates among those women who participated in MRI screening with those who did not (Lubinski et al). After a median follow-up of 9.2 years, 344 women (13.8%) developed breast cancer, and 35 (1.4%) died from breast cancer. There was an 80% reduction in breast cancer mortality among BRCA1 mutation carriers who participated in MRI surveillance vs those who did not (age-adjusted hazard ratio [HR] 0.20; 95% CI 0.10-0.43; P < .001), but this was not observed for women with BRCA2 mutations (age-adjusted HR 0.87; 95% CI 0.10-17.25; P = .93). At 20 years, the breast cancer mortality rate was 3.2% in the MRI surveillance group compared with 14.9% in the group who did not undergo surveillance. A separate cohort study from Ontario, Canada, including 489 women with BRCA1/2 pathogenic mutations found a 2.0% rate of breast cancer-related mortality at 20 years after the first MRI screening.[3] These data support an intensified surveillance schedule for BRCA mutation carriers, with a need for further research and insight in the BRCA2 population.

A positive family history of cancer and obesity are established risk factors for development of breast cancer among women.[4,5] A population-based cohort study that included 15,055 Chinese women evaluated the association and interaction between body mass index (BMI) and family history of cancer on the risk for breast cancer (Cao et al). The incidence risk for breast cancer was highest in the group with obesity vs the group with normal weight (adjusted HR 2.09; 95% CI 1.42-3.07), and those with a family history of cancer also had an increased risk vs those without a family history of cancer (adjusted HR 1.63; 95% CI 1.22-2.49). Furthermore, women with a BMI ≥ 24 and family history of cancer had a higher risk for breast cancer development compared with women with a BMI < 24 and no family history of cancer (adjusted HR 2.06; 95% CI 1.39-3.06). This study indicates a heightened breast cancer risk when cancer family history and obesity coexist, suggesting the importance of addressing modifiable risk factors and targeting lifestyle interventions in this population.

Triple-negative breast cancer (TNBC), although exhibiting its own heterogeneity, has various features that differentiate this subtype from luminal breast cancers. For example, TNBC generally has a more aggressive course, increased responsiveness to chemotherapy, and earlier pattern of recurrence compared with hormone receptor–positive disease. Prior studies have also shown that established breast cancer risk factors reflect those for the luminal A subtype, whereas those for TNBC are less consistent.[6] A meta-analysis that included 33 studies evaluated the association between traditional breast cancer risk factors and TNBC incidence (Kumar et al). Family history (odds ratio [OR] 1.55; 95% CI 1.34-1.81; P < .001), longer duration of oral contraceptive use (OR 1.29; 95% CI 1.08-1.55; P < .001), and higher breast density (OR 2.19; 95% CI 1.67-2.88; P < .001) were significantly associated with an increased risk for TNBC. Factors including later age at menarche, later age at first birth, and breastfeeding were associated with reduced risk for TNBC. Furthermore, there was no significant association with parity, menopausal hormone therapy, alcohol, smoking, and BMI. This study highlights distinct risk factors that may contribute to a higher risk for TNBC, and future research will be valuable to better elucidate the mechanisms at play and to further understand the differences within this subtype itself.

Additional References

  1. National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic. Version 3.2024. Source 
  2. Saadatmand S, Geuzinge HA, Rutgers EJT, et al; on behalf of the FaMRIsc study group. MRI versus mammography for breast cancer screening in women with familial risk (FaMRIsc): A multicentre, randomised, controlled trial. Lancet Oncol. 2019;20:1136-1147. doi: 10.1016/S1470-2045(19)30275-X  Source
  3. Warner E, Zhu S, Plewes DB, et al. Breast cancer mortality among women with a BRCA1 or BRCA2 mutation in a magnetic resonance imaging plus mammography screening program. Cancers (Basel). 2020;12:3479. doi: 10.3390/cancers12113479 Source
  4. Picon-Ruiz M, Morata-Tarifa C, Valle-Goffin JJ, et al. Obesity and adverse breast cancer risk and outcome: Mechanistic insights and strategies for intervention. CA Cancer J Clin. 2017;67:378-397. doi: 10.3322/caac.21405 Source
  5. Engmann NJ, Golmakani MK, Miglioretti DL, et al; for the Breast Cancer Surveillance Consortium. Population-attributable risk proportion of clinical risk factors for breast cancer. JAMA Oncol. 2017;3:1228-1236. doi: 10.1001/jamaoncol.2016.6326 Source
  6. Barnard ME, Boeke CE, Tamimi RM. Established breast cancer risk factors and risk of intrinsic tumor subtypes. Biochim Biophys Acta Rev Cancer. 2015;1856:73-85. doi: 10.1016/j.bbcan.2015.0002 Source
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
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
Activity Salesforce Deliverable ID
398326.1
Activity ID
109750
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Choosing Treatments of AD, and Possible Connection to Learning Issues, April 2024

Article Type
Changed
Wed, 04/10/2024 - 09:13
Dr. Feldman scans the journals, so you don’t have to!

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
I'm usually complaining that studies that show "a significant increased risk" for something are usually finding an insignificant risk that is statistically significant but not clinically meaningful. Not this time! The study by Ma and colleagues found that 11% of children with atopic dermatitis (AD) and about 6% of children without AD experienced difficulties with learning. This is a big difference. It means that 1 in 16 children without AD have difficulties with learning and that about 1 in 9 with AD have difficulties with learning. I think that means if you see 20 children with AD, 1 will have learning difficulties due to the AD. This is not surprising. AD has big effects on patients' lives. Sleep disturbance and difficulty concentrating might cause the learning difficulties. On the other hand, it's also possible that the findings could be confounded by people with AD being more likely to be diagnosed as having learning difficulties even when the rate of learning difficulties is the same.


Not everyone with AD treated with dupilumab gets clear or almost clear in clinical trials. The study by Cork and colleagues looked to see whether those patients who did not get to clear or almost clear were still having clinically meaningful improvement. To test this, the investigators looked at patients who still had mild or worse disease and then at the proportion of those patients at week 16 who achieved a composite endpoint encompassing clinically meaningful changes in AD signs, symptoms, and quality of life: ≥50% improvement in Eczema Area and Severity Index or ≥4-point reduction in worst scratch/itch numerical rating scale, or ≥6-point reduction in Children's Dermatology Life Quality Index/Infants' Dermatitis Quality of Life Index. Significantly more patients, both clinically and statistically significantly more, receiving dupilumab vs placebo achieved the composite endpoint (77.7% vs 24.6%; P < .0001). 


The "success rate" reported in clinical trials underestimates how often patients can be successfully treated with dupilumab. I don't need a complicated composite outcome to know this. I just use the standardized 2-point Patient Global Assessment measure. I ask patients, "How are you doing?" If they say "Great," that's success. If they say, "Not so good," that's failure. I think about 80% of patients with AD treated with dupilumab have success based on this standard.


Hand dermatitis can be quite resistant to treatment. Even making a diagnosis can be challenging, as psoriasis and dermatitis of the hands looks so similar to me (and when I used to send biopsies and ask the pathologist whether it's dermatitis or psoriasis, invariably the dermatopathologist responded "yes"). The study by Kamphuis and colleagues examined the efficacy of abrocitinib in just over 100 patients with hand eczema who were enrolled in the BioDay registry. Such registries are very helpful for assessing real-world results. The drug seemed reasonably successful, with only about 30% discontinuing treatment. About two thirds of the discontinuations were due to inefficacy and about one third to an adverse event.


I think there's real value in prescribing the treatments patients want. Studies like the one by Ameen and colleagues, using a discrete-choice methodology, allows one to determine patients' average preferences. In this study, the discrete-choice approach found that patients prefer safety over other attributes. Some years ago, my colleagues and I queried patients to get a sense of their quantitative preferences for different treatments. Our study also found that patients preferred safety over other attributes. However, when we asked them to choose among different treatment options, they didn't choose the safest one. I think they believe that they prefer safety, but I'm not sure they really do. In any case, the average preference of the entire population of people with AD isn't really all that important when we've got just one patient sitting in front of us. It's that particular patient's preference that should drive the treatment plan.

Author and Disclosure Information

Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

He has reported no disclosures.

Publications
Topics
Sections
Author and Disclosure Information

Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

He has reported no disclosures.

Author and Disclosure Information

Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

He has reported no disclosures.

Dr. Feldman scans the journals, so you don’t have to!
Dr. Feldman scans the journals, so you don’t have to!

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
I'm usually complaining that studies that show "a significant increased risk" for something are usually finding an insignificant risk that is statistically significant but not clinically meaningful. Not this time! The study by Ma and colleagues found that 11% of children with atopic dermatitis (AD) and about 6% of children without AD experienced difficulties with learning. This is a big difference. It means that 1 in 16 children without AD have difficulties with learning and that about 1 in 9 with AD have difficulties with learning. I think that means if you see 20 children with AD, 1 will have learning difficulties due to the AD. This is not surprising. AD has big effects on patients' lives. Sleep disturbance and difficulty concentrating might cause the learning difficulties. On the other hand, it's also possible that the findings could be confounded by people with AD being more likely to be diagnosed as having learning difficulties even when the rate of learning difficulties is the same.


Not everyone with AD treated with dupilumab gets clear or almost clear in clinical trials. The study by Cork and colleagues looked to see whether those patients who did not get to clear or almost clear were still having clinically meaningful improvement. To test this, the investigators looked at patients who still had mild or worse disease and then at the proportion of those patients at week 16 who achieved a composite endpoint encompassing clinically meaningful changes in AD signs, symptoms, and quality of life: ≥50% improvement in Eczema Area and Severity Index or ≥4-point reduction in worst scratch/itch numerical rating scale, or ≥6-point reduction in Children's Dermatology Life Quality Index/Infants' Dermatitis Quality of Life Index. Significantly more patients, both clinically and statistically significantly more, receiving dupilumab vs placebo achieved the composite endpoint (77.7% vs 24.6%; P < .0001). 


The "success rate" reported in clinical trials underestimates how often patients can be successfully treated with dupilumab. I don't need a complicated composite outcome to know this. I just use the standardized 2-point Patient Global Assessment measure. I ask patients, "How are you doing?" If they say "Great," that's success. If they say, "Not so good," that's failure. I think about 80% of patients with AD treated with dupilumab have success based on this standard.


Hand dermatitis can be quite resistant to treatment. Even making a diagnosis can be challenging, as psoriasis and dermatitis of the hands looks so similar to me (and when I used to send biopsies and ask the pathologist whether it's dermatitis or psoriasis, invariably the dermatopathologist responded "yes"). The study by Kamphuis and colleagues examined the efficacy of abrocitinib in just over 100 patients with hand eczema who were enrolled in the BioDay registry. Such registries are very helpful for assessing real-world results. The drug seemed reasonably successful, with only about 30% discontinuing treatment. About two thirds of the discontinuations were due to inefficacy and about one third to an adverse event.


I think there's real value in prescribing the treatments patients want. Studies like the one by Ameen and colleagues, using a discrete-choice methodology, allows one to determine patients' average preferences. In this study, the discrete-choice approach found that patients prefer safety over other attributes. Some years ago, my colleagues and I queried patients to get a sense of their quantitative preferences for different treatments. Our study also found that patients preferred safety over other attributes. However, when we asked them to choose among different treatment options, they didn't choose the safest one. I think they believe that they prefer safety, but I'm not sure they really do. In any case, the average preference of the entire population of people with AD isn't really all that important when we've got just one patient sitting in front of us. It's that particular patient's preference that should drive the treatment plan.

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
I'm usually complaining that studies that show "a significant increased risk" for something are usually finding an insignificant risk that is statistically significant but not clinically meaningful. Not this time! The study by Ma and colleagues found that 11% of children with atopic dermatitis (AD) and about 6% of children without AD experienced difficulties with learning. This is a big difference. It means that 1 in 16 children without AD have difficulties with learning and that about 1 in 9 with AD have difficulties with learning. I think that means if you see 20 children with AD, 1 will have learning difficulties due to the AD. This is not surprising. AD has big effects on patients' lives. Sleep disturbance and difficulty concentrating might cause the learning difficulties. On the other hand, it's also possible that the findings could be confounded by people with AD being more likely to be diagnosed as having learning difficulties even when the rate of learning difficulties is the same.


Not everyone with AD treated with dupilumab gets clear or almost clear in clinical trials. The study by Cork and colleagues looked to see whether those patients who did not get to clear or almost clear were still having clinically meaningful improvement. To test this, the investigators looked at patients who still had mild or worse disease and then at the proportion of those patients at week 16 who achieved a composite endpoint encompassing clinically meaningful changes in AD signs, symptoms, and quality of life: ≥50% improvement in Eczema Area and Severity Index or ≥4-point reduction in worst scratch/itch numerical rating scale, or ≥6-point reduction in Children's Dermatology Life Quality Index/Infants' Dermatitis Quality of Life Index. Significantly more patients, both clinically and statistically significantly more, receiving dupilumab vs placebo achieved the composite endpoint (77.7% vs 24.6%; P < .0001). 


The "success rate" reported in clinical trials underestimates how often patients can be successfully treated with dupilumab. I don't need a complicated composite outcome to know this. I just use the standardized 2-point Patient Global Assessment measure. I ask patients, "How are you doing?" If they say "Great," that's success. If they say, "Not so good," that's failure. I think about 80% of patients with AD treated with dupilumab have success based on this standard.


Hand dermatitis can be quite resistant to treatment. Even making a diagnosis can be challenging, as psoriasis and dermatitis of the hands looks so similar to me (and when I used to send biopsies and ask the pathologist whether it's dermatitis or psoriasis, invariably the dermatopathologist responded "yes"). The study by Kamphuis and colleagues examined the efficacy of abrocitinib in just over 100 patients with hand eczema who were enrolled in the BioDay registry. Such registries are very helpful for assessing real-world results. The drug seemed reasonably successful, with only about 30% discontinuing treatment. About two thirds of the discontinuations were due to inefficacy and about one third to an adverse event.


I think there's real value in prescribing the treatments patients want. Studies like the one by Ameen and colleagues, using a discrete-choice methodology, allows one to determine patients' average preferences. In this study, the discrete-choice approach found that patients prefer safety over other attributes. Some years ago, my colleagues and I queried patients to get a sense of their quantitative preferences for different treatments. Our study also found that patients preferred safety over other attributes. However, when we asked them to choose among different treatment options, they didn't choose the safest one. I think they believe that they prefer safety, but I'm not sure they really do. In any case, the average preference of the entire population of people with AD isn't really all that important when we've got just one patient sitting in front of us. It's that particular patient's preference that should drive the treatment plan.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 03/26/2024 - 15:00
Un-Gate On Date
Tue, 03/26/2024 - 15:00
Use ProPublica
CFC Schedule Remove Status
Tue, 03/26/2024 - 15:00
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
Activity Salesforce Deliverable ID
400680.1
Activity ID
110501
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
RINVOQ [ 5260 ]

Commentary: PsA Comorbidities and Treatment Safety and Effectiveness, March 2024

Article Type
Changed
Wed, 02/28/2024 - 13:54
Dr. Chandran scans the journals, so you don't have to!

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20lang%3D%22EN-CA%22%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Studies published over the past month have focused on treatment of psoriatic arthritis (PsA) as well as comorbidities. Using the resources of the Rochester Epidemiology Project, Karmacharya and colleagues demonstrated that comorbidities, especially multimorbidity (presence of two or more comorbidities), are strong risk factors for the development of PsA in patients with psoriasis. In this retrospective cohort study that included 817 patients with incident psoriasis and 849 age- and sex-matched controls without psoriasis, researchers showed that the cumulative incidence of PsA in patients with psoriasis was low, but the risk for PsA was threefold higher in those with multimorbidity. Thus, patients with multimorbid psoriasis should be monitored for the potential development of PsA.

 

An important comorbidity of PsA is vascular inflammation leading to accelerated atherosclerosis, and higher risk for cardiovascular and cerebrovascular disease. Previously, vascular imaging modalities have demonstrated vascular inflammation in PsA. In a cross-sectional study that included 75 patients with active PsA and 40 control individuals without PsA, Kleinrensink and colleagues demonstrated that vascular inflammation of the whole aorta was significantly increased in patients with PsA vs control individuals. Of note, the association remained significant after adjusting for gender, age, body mass index, mean arterial pressure, and aortic calcification, but it was not associated with disease-related parameters. Further studies to determine the contributions of PsA per se and its comorbidities to vascular inflammation are required. Nevertheless, the management of PsA should include close monitoring and aggressive treatment of risk factors for atherosclerotic vascular disease.

 

Psychotic disorders are known to be associated with psoriasis, but their association with PsA is less well known. Using French health administrative data, Brenaut and colleagues showed that the prevalence of psychotic disorders was higher in individuals with psoriasis but surprisingly lower in individuals with PsA, compared with the general population. Moreover, a co-diagnosis of psoriasis/PsA and psychotic disorders was associated with an increased mortality rate and at a lower age.

 

Clinical trials have demonstrated that Janus kinase (JAK) inhibitors have a remarkable efficacy in the treatment of the musculoskeletal manifestations of PsA. Observational studies are important to evaluate effectiveness in real-world settings. In a study that included 123 patients with PsA from the CorEvitas PsA/Spondyloarthritis Registry who were treated with tofacitinib, Mease and colleagues observed that a quarter of patients achieved a state of low disease activity, based on the Clinical Disease Activity Index for PsA at 6 ± 3 months of follow-up. A substantial proportion of patients also reported the resolution of dactylitis (29.4%) and enthesitis (42.9%). Although these results are remarkable compared with what was seen with older therapies, one must note that only a quarter of patients achieved remission; more effective regimens for improving outcomes in PsA are required.

 

The safety of newer therapies is always of concern. It is reassuring that a meta-analysis of six randomized controlled trials that included 5038 patients with PsA who received either risankizumab (an anti-interleukin-23 antibody) or placebo by Su and colleagues demonstrated that the incidences of serious adverse events and serious treatment-emergent adverse events were similar between the risankizumab and placebo groups. Given the excellent safety profile of some of the newer therapies for PsA, trials with combinations of newer targeted therapies in treatment-resistant PsA should be conducted.

 

 

Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Publications
Topics
Sections
Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Author and Disclosure Information

Vinod Chandran MBBS, MD, DM, PhD, FRCPC

Staff Physician, Department of Medicine/Rheumatology, University Health Network, Toronto, ON, Canada

Vinod Chandran, MBBS, MD, DM, PhD, has disclosed the following relevant financial relationships: Member of the board of directors of the Group for Research and Assessment of Psoriasis and Psoriatic Arthritis (GRAPPA). Received research grant from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly. Received income in an amount equal to or greater than $250 from: Amgen; AbbVie; Bristol-Myers Squibb; Eli Lilly; Janssen; Novartis; UCB.
Spousal employment: AstraZeneca

Dr. Chandran scans the journals, so you don't have to!
Dr. Chandran scans the journals, so you don't have to!

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20lang%3D%22EN-CA%22%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Studies published over the past month have focused on treatment of psoriatic arthritis (PsA) as well as comorbidities. Using the resources of the Rochester Epidemiology Project, Karmacharya and colleagues demonstrated that comorbidities, especially multimorbidity (presence of two or more comorbidities), are strong risk factors for the development of PsA in patients with psoriasis. In this retrospective cohort study that included 817 patients with incident psoriasis and 849 age- and sex-matched controls without psoriasis, researchers showed that the cumulative incidence of PsA in patients with psoriasis was low, but the risk for PsA was threefold higher in those with multimorbidity. Thus, patients with multimorbid psoriasis should be monitored for the potential development of PsA.

 

An important comorbidity of PsA is vascular inflammation leading to accelerated atherosclerosis, and higher risk for cardiovascular and cerebrovascular disease. Previously, vascular imaging modalities have demonstrated vascular inflammation in PsA. In a cross-sectional study that included 75 patients with active PsA and 40 control individuals without PsA, Kleinrensink and colleagues demonstrated that vascular inflammation of the whole aorta was significantly increased in patients with PsA vs control individuals. Of note, the association remained significant after adjusting for gender, age, body mass index, mean arterial pressure, and aortic calcification, but it was not associated with disease-related parameters. Further studies to determine the contributions of PsA per se and its comorbidities to vascular inflammation are required. Nevertheless, the management of PsA should include close monitoring and aggressive treatment of risk factors for atherosclerotic vascular disease.

 

Psychotic disorders are known to be associated with psoriasis, but their association with PsA is less well known. Using French health administrative data, Brenaut and colleagues showed that the prevalence of psychotic disorders was higher in individuals with psoriasis but surprisingly lower in individuals with PsA, compared with the general population. Moreover, a co-diagnosis of psoriasis/PsA and psychotic disorders was associated with an increased mortality rate and at a lower age.

 

Clinical trials have demonstrated that Janus kinase (JAK) inhibitors have a remarkable efficacy in the treatment of the musculoskeletal manifestations of PsA. Observational studies are important to evaluate effectiveness in real-world settings. In a study that included 123 patients with PsA from the CorEvitas PsA/Spondyloarthritis Registry who were treated with tofacitinib, Mease and colleagues observed that a quarter of patients achieved a state of low disease activity, based on the Clinical Disease Activity Index for PsA at 6 ± 3 months of follow-up. A substantial proportion of patients also reported the resolution of dactylitis (29.4%) and enthesitis (42.9%). Although these results are remarkable compared with what was seen with older therapies, one must note that only a quarter of patients achieved remission; more effective regimens for improving outcomes in PsA are required.

 

The safety of newer therapies is always of concern. It is reassuring that a meta-analysis of six randomized controlled trials that included 5038 patients with PsA who received either risankizumab (an anti-interleukin-23 antibody) or placebo by Su and colleagues demonstrated that the incidences of serious adverse events and serious treatment-emergent adverse events were similar between the risankizumab and placebo groups. Given the excellent safety profile of some of the newer therapies for PsA, trials with combinations of newer targeted therapies in treatment-resistant PsA should be conducted.

 

 

vinod-chandran-2_0.jpg
%3Cp%3E%3Cspan%20lang%3D%22EN-CA%22%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EVinod%20Chandran%2C%20MBBS%2C%20MD%2C%20DM%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Studies published over the past month have focused on treatment of psoriatic arthritis (PsA) as well as comorbidities. Using the resources of the Rochester Epidemiology Project, Karmacharya and colleagues demonstrated that comorbidities, especially multimorbidity (presence of two or more comorbidities), are strong risk factors for the development of PsA in patients with psoriasis. In this retrospective cohort study that included 817 patients with incident psoriasis and 849 age- and sex-matched controls without psoriasis, researchers showed that the cumulative incidence of PsA in patients with psoriasis was low, but the risk for PsA was threefold higher in those with multimorbidity. Thus, patients with multimorbid psoriasis should be monitored for the potential development of PsA.

 

An important comorbidity of PsA is vascular inflammation leading to accelerated atherosclerosis, and higher risk for cardiovascular and cerebrovascular disease. Previously, vascular imaging modalities have demonstrated vascular inflammation in PsA. In a cross-sectional study that included 75 patients with active PsA and 40 control individuals without PsA, Kleinrensink and colleagues demonstrated that vascular inflammation of the whole aorta was significantly increased in patients with PsA vs control individuals. Of note, the association remained significant after adjusting for gender, age, body mass index, mean arterial pressure, and aortic calcification, but it was not associated with disease-related parameters. Further studies to determine the contributions of PsA per se and its comorbidities to vascular inflammation are required. Nevertheless, the management of PsA should include close monitoring and aggressive treatment of risk factors for atherosclerotic vascular disease.

 

Psychotic disorders are known to be associated with psoriasis, but their association with PsA is less well known. Using French health administrative data, Brenaut and colleagues showed that the prevalence of psychotic disorders was higher in individuals with psoriasis but surprisingly lower in individuals with PsA, compared with the general population. Moreover, a co-diagnosis of psoriasis/PsA and psychotic disorders was associated with an increased mortality rate and at a lower age.

 

Clinical trials have demonstrated that Janus kinase (JAK) inhibitors have a remarkable efficacy in the treatment of the musculoskeletal manifestations of PsA. Observational studies are important to evaluate effectiveness in real-world settings. In a study that included 123 patients with PsA from the CorEvitas PsA/Spondyloarthritis Registry who were treated with tofacitinib, Mease and colleagues observed that a quarter of patients achieved a state of low disease activity, based on the Clinical Disease Activity Index for PsA at 6 ± 3 months of follow-up. A substantial proportion of patients also reported the resolution of dactylitis (29.4%) and enthesitis (42.9%). Although these results are remarkable compared with what was seen with older therapies, one must note that only a quarter of patients achieved remission; more effective regimens for improving outcomes in PsA are required.

 

The safety of newer therapies is always of concern. It is reassuring that a meta-analysis of six randomized controlled trials that included 5038 patients with PsA who received either risankizumab (an anti-interleukin-23 antibody) or placebo by Su and colleagues demonstrated that the incidences of serious adverse events and serious treatment-emergent adverse events were similar between the risankizumab and placebo groups. Given the excellent safety profile of some of the newer therapies for PsA, trials with combinations of newer targeted therapies in treatment-resistant PsA should be conducted.

 

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 04/05/2021 - 09:15
Un-Gate On Date
Mon, 04/05/2021 - 09:15
Use ProPublica
CFC Schedule Remove Status
Mon, 04/05/2021 - 09:15
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
Activity Salesforce Deliverable ID
400312.1
Activity ID
110008
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
SKYRIZI [ 5052 ]

Commentary: New Research on BC Chemotherapies, March 2024

Article Type
Changed
Wed, 03/13/2024 - 14:30
Dr Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Adjuvant endocrine therapy with aromatase inhibitors (AI) significantly reduces risk for recurrence and favorably affects survival outcomes in early breast cancer (EBC). However, these therapies also carry potential side effects due to estrogen depletion, including increased bone resorption, decreased bone mineral density, and subsequently an increased risk for fracture. Bone-strengthening agents can help mitigate the risk for AI-related bone loss. Studies have demonstrated a benefit in terms of breast cancer outcomes with their use in the adjuvant setting.[1] A prospective, single-center cohort study that included 237 postmenopausal patients with hormone receptor–positive EBC who were receiving adjuvant AI treatment and denosumab every 6 months explored the association between risk factors for bone fracture and vertebral fracture progression in this population (Cosentini et al). After 18 months of treatment, a total of 4.4% of patients were noted to have vertebral fracture progression. Both Fracture Risk Assessment Tool (FRAX) score (odds ratio [OR] 3.95; 95% CI 1.09-14.39; P = .04) and percentage of fat body mass  (OR 5.41; 95% CI 1.49-19.59; P = .01) were independent variables associated with vertebral fracture progression. The relationship between obesity and bone density is complex; higher estrogen levels derived from adipose tissue exert a protective effect on bone mineral density, whereas inflammatory cytokines and other hormones can adversely affect bone quality.[2] This study highlights the beneficial role of modalities such as diet and exercise to achieve healthy body mass, and how these can work in conjunction with antiresorptive therapy (such as denosumab) to benefit bone health in women receiving AI.

The phase 3 KEYNOTE-355 trial established the role of chemotherapy in combination with pembrolizumab in the first-line setting for programmed death-ligand 1 (PD-L1)–positive advanced triple-negative breast cancer (TNBC). Patients unselected for PD-L1 status in this trial who received platinum- or taxane-based chemotherapy with placebo had a median progression-free survival of 5.6 months.[3] Strategies to improve upon efficacy and tolerability are desired in this space, and various trials have evaluated "switch maintenance" that involves receipt of an intensive induction regimen followed by a switch to an alternative/more tolerable regimen after response is achieved.[4] The phase II DORA trial randomized 45 patients with advanced TNBC and ongoing stable disease or complete or partial response from first- or second-line platinum-based chemotherapy to a maintenance regimen of olaparib (300 mg orally twice daily) with or without durvalumab (1500 mg on day 1 and every 4 weeks) (Tan et al). At a median follow-up of 9.8 months, median progression-free survival was 4.0 months (95% CI 2.6-6.1) with olaparib and 6.1 months (95% CI 3.7-10.1) with the combination; both were significantly longer than the historical control of continued platinum-based therapy (P = .0023 and P < .0001, respectively). Durable disease control appeared more pronounced in patients with complete or partial response to prior platinum therapy, and no new safety signals were observed. Future efforts to study this approach include the phase 2/3 KEYLYNK-009 trial, which is evaluating olaparib plus pembrolizumab maintenance therapy after first-line chemotherapy plus pembrolizumab for TNBC.[5]

TNBC is a heterogenous subtype, characterized by aggressive biology, and it benefits from chemotherapy and immunotherapy treatment approaches. Presently, the management of early-stage TNBC often involves neoadjuvant systemic therapy; however, a proportion of patients receive treatment in the postoperative setting, highlighting the relevance of time to initiation of adjuvant therapy as well.[6] Various prior studies have showed that delayed administration of adjuvant chemotherapy for EBC can lead to adverse survival outcomes. Furthermore, this effect is subtype-dependent, with more aggressive tumors (luminal B, triple-negative, human epidermal growth factor receptor 2 [HER2]-positive) exhibiting inferior outcomes with delayed chemotherapy.[7] A retrospective cohort study that included 245 patients with early TNBC who received adjuvant chemotherapy after surgery evaluated the impact of time to initiation of adjuvant therapy in this population (Hatzipanagiotou et al). Superior survival outcomes were observed for the group receiving systemic therapy 22-28 days after surgery (median overall survival 10.2 years) compared with those receiving adjuvant chemotherapy at later time points (29-35 days, 36-42 days, and >6 weeks after surgery; median overall survival 8.3 years, 7.8 years, and 6.9 years, respectively). Patients receiving chemotherapy 22-28 days after surgery had significantly better survival than those receiving chemotherapy 29-35 days (P = .043) and >6 weeks (P = 0.033) postoperatively. This study emphasizes the importance of timely administration of adjuvant chemotherapy for early TNBC, and efforts aimed to identify potential challenges and propose solutions to optimize outcomes in this space are valuable.

Additional References

  1. Gnant M, Frantal S, Pfeiler G, et al, for the Austrian Breast & Colorectal Cancer Study Group. Long-term outcomes of adjuvant denosumab in breast cancer. NEJM Evid. 2022;1:EVIDoa2200162. doi: 10.1056/EVIDoa2200162 Source
  2. Fassio A, Idolazzi L, Rossini M, et al. The obesity paradox and osteoporosis. Eating and Weight Disorders - Studies on Anorexia, Bulimia and Obesity. 2018;23:293-30 doi: 10.1007/s40519-018-0505-2 Source
  3. Cortes J, Cescon DW, Rugo HS, et al, for the KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:1817-1828. doi: 10.1016/S0140-6736(20)32531-9 Source
  4. Bachelot T, Filleron T, Bieche I, et al. Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: The randomized phase II SAFIR02-BREAST IMMUNO trial. Nat Med. 2021;27:250-255. doi: 10.1038/s41591-020-01189-2 Source
  5. Saji S, Cussac AL, Andre F, et al. 68TiP KEYLYNK-009: a phase II/III, open-label, randomized study of pembrolizumab (pembro) + olaparib (ola) vs pembro + chemotherapy after induction with first-line (1L) pembro + chemo in patients (pts) with locally recurrent inoperable or metastatic TNBC (abstract). Ann Oncol. 2020;31(Suppl 6):S1268. doi: 10.1016/j.annonc.2020.10.088 Source
  6. Ortmann O, Blohmer JU, Sibert NT, et al for 55 breast cancer centers certified by the German Cancer Society. Current clinical practice and outcome of neoadjuvant chemotherapy for early breast cancer: Analysis of individual data from 94,638 patients treated in 55 breast cancer centers. J Cancer Res Clin Oncol. 2023;149:1195-1209. doi: 10.1007/s00432-022-03938-x Source
  7. Yu KD, Fan L, Qiu LX, et al. Influence of delayed initiation of adjuvant chemotherapy on breast cancer survival is subtype-dependent. Oncotarget. 2017;8:46549-46556. doi: 10.18632/oncotarget.10551 Source
Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin Roesch, MD Assistant Professor, Department of Medicine, Cleveland Clinic Lerner College of Medicine; Assocaite Staff Physician, Hematology and Oncology, Cancer Institute, Cleveland, Ohio

Erin E. Roesch, MD, has disclosed the following relevant financial relationships: Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr Roesch scans the journals so you don't have to!
Dr Roesch scans the journals so you don't have to!

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Adjuvant endocrine therapy with aromatase inhibitors (AI) significantly reduces risk for recurrence and favorably affects survival outcomes in early breast cancer (EBC). However, these therapies also carry potential side effects due to estrogen depletion, including increased bone resorption, decreased bone mineral density, and subsequently an increased risk for fracture. Bone-strengthening agents can help mitigate the risk for AI-related bone loss. Studies have demonstrated a benefit in terms of breast cancer outcomes with their use in the adjuvant setting.[1] A prospective, single-center cohort study that included 237 postmenopausal patients with hormone receptor–positive EBC who were receiving adjuvant AI treatment and denosumab every 6 months explored the association between risk factors for bone fracture and vertebral fracture progression in this population (Cosentini et al). After 18 months of treatment, a total of 4.4% of patients were noted to have vertebral fracture progression. Both Fracture Risk Assessment Tool (FRAX) score (odds ratio [OR] 3.95; 95% CI 1.09-14.39; P = .04) and percentage of fat body mass  (OR 5.41; 95% CI 1.49-19.59; P = .01) were independent variables associated with vertebral fracture progression. The relationship between obesity and bone density is complex; higher estrogen levels derived from adipose tissue exert a protective effect on bone mineral density, whereas inflammatory cytokines and other hormones can adversely affect bone quality.[2] This study highlights the beneficial role of modalities such as diet and exercise to achieve healthy body mass, and how these can work in conjunction with antiresorptive therapy (such as denosumab) to benefit bone health in women receiving AI.

The phase 3 KEYNOTE-355 trial established the role of chemotherapy in combination with pembrolizumab in the first-line setting for programmed death-ligand 1 (PD-L1)–positive advanced triple-negative breast cancer (TNBC). Patients unselected for PD-L1 status in this trial who received platinum- or taxane-based chemotherapy with placebo had a median progression-free survival of 5.6 months.[3] Strategies to improve upon efficacy and tolerability are desired in this space, and various trials have evaluated "switch maintenance" that involves receipt of an intensive induction regimen followed by a switch to an alternative/more tolerable regimen after response is achieved.[4] The phase II DORA trial randomized 45 patients with advanced TNBC and ongoing stable disease or complete or partial response from first- or second-line platinum-based chemotherapy to a maintenance regimen of olaparib (300 mg orally twice daily) with or without durvalumab (1500 mg on day 1 and every 4 weeks) (Tan et al). At a median follow-up of 9.8 months, median progression-free survival was 4.0 months (95% CI 2.6-6.1) with olaparib and 6.1 months (95% CI 3.7-10.1) with the combination; both were significantly longer than the historical control of continued platinum-based therapy (P = .0023 and P < .0001, respectively). Durable disease control appeared more pronounced in patients with complete or partial response to prior platinum therapy, and no new safety signals were observed. Future efforts to study this approach include the phase 2/3 KEYLYNK-009 trial, which is evaluating olaparib plus pembrolizumab maintenance therapy after first-line chemotherapy plus pembrolizumab for TNBC.[5]

TNBC is a heterogenous subtype, characterized by aggressive biology, and it benefits from chemotherapy and immunotherapy treatment approaches. Presently, the management of early-stage TNBC often involves neoadjuvant systemic therapy; however, a proportion of patients receive treatment in the postoperative setting, highlighting the relevance of time to initiation of adjuvant therapy as well.[6] Various prior studies have showed that delayed administration of adjuvant chemotherapy for EBC can lead to adverse survival outcomes. Furthermore, this effect is subtype-dependent, with more aggressive tumors (luminal B, triple-negative, human epidermal growth factor receptor 2 [HER2]-positive) exhibiting inferior outcomes with delayed chemotherapy.[7] A retrospective cohort study that included 245 patients with early TNBC who received adjuvant chemotherapy after surgery evaluated the impact of time to initiation of adjuvant therapy in this population (Hatzipanagiotou et al). Superior survival outcomes were observed for the group receiving systemic therapy 22-28 days after surgery (median overall survival 10.2 years) compared with those receiving adjuvant chemotherapy at later time points (29-35 days, 36-42 days, and >6 weeks after surgery; median overall survival 8.3 years, 7.8 years, and 6.9 years, respectively). Patients receiving chemotherapy 22-28 days after surgery had significantly better survival than those receiving chemotherapy 29-35 days (P = .043) and >6 weeks (P = 0.033) postoperatively. This study emphasizes the importance of timely administration of adjuvant chemotherapy for early TNBC, and efforts aimed to identify potential challenges and propose solutions to optimize outcomes in this space are valuable.

Additional References

  1. Gnant M, Frantal S, Pfeiler G, et al, for the Austrian Breast & Colorectal Cancer Study Group. Long-term outcomes of adjuvant denosumab in breast cancer. NEJM Evid. 2022;1:EVIDoa2200162. doi: 10.1056/EVIDoa2200162 Source
  2. Fassio A, Idolazzi L, Rossini M, et al. The obesity paradox and osteoporosis. Eating and Weight Disorders - Studies on Anorexia, Bulimia and Obesity. 2018;23:293-30 doi: 10.1007/s40519-018-0505-2 Source
  3. Cortes J, Cescon DW, Rugo HS, et al, for the KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:1817-1828. doi: 10.1016/S0140-6736(20)32531-9 Source
  4. Bachelot T, Filleron T, Bieche I, et al. Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: The randomized phase II SAFIR02-BREAST IMMUNO trial. Nat Med. 2021;27:250-255. doi: 10.1038/s41591-020-01189-2 Source
  5. Saji S, Cussac AL, Andre F, et al. 68TiP KEYLYNK-009: a phase II/III, open-label, randomized study of pembrolizumab (pembro) + olaparib (ola) vs pembro + chemotherapy after induction with first-line (1L) pembro + chemo in patients (pts) with locally recurrent inoperable or metastatic TNBC (abstract). Ann Oncol. 2020;31(Suppl 6):S1268. doi: 10.1016/j.annonc.2020.10.088 Source
  6. Ortmann O, Blohmer JU, Sibert NT, et al for 55 breast cancer centers certified by the German Cancer Society. Current clinical practice and outcome of neoadjuvant chemotherapy for early breast cancer: Analysis of individual data from 94,638 patients treated in 55 breast cancer centers. J Cancer Res Clin Oncol. 2023;149:1195-1209. doi: 10.1007/s00432-022-03938-x Source
  7. Yu KD, Fan L, Qiu LX, et al. Influence of delayed initiation of adjuvant chemotherapy on breast cancer survival is subtype-dependent. Oncotarget. 2017;8:46549-46556. doi: 10.18632/oncotarget.10551 Source

roesch_erin_headshot_1_0_0_0_0.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3EErin%20Roesch%2C%20MD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Adjuvant endocrine therapy with aromatase inhibitors (AI) significantly reduces risk for recurrence and favorably affects survival outcomes in early breast cancer (EBC). However, these therapies also carry potential side effects due to estrogen depletion, including increased bone resorption, decreased bone mineral density, and subsequently an increased risk for fracture. Bone-strengthening agents can help mitigate the risk for AI-related bone loss. Studies have demonstrated a benefit in terms of breast cancer outcomes with their use in the adjuvant setting.[1] A prospective, single-center cohort study that included 237 postmenopausal patients with hormone receptor–positive EBC who were receiving adjuvant AI treatment and denosumab every 6 months explored the association between risk factors for bone fracture and vertebral fracture progression in this population (Cosentini et al). After 18 months of treatment, a total of 4.4% of patients were noted to have vertebral fracture progression. Both Fracture Risk Assessment Tool (FRAX) score (odds ratio [OR] 3.95; 95% CI 1.09-14.39; P = .04) and percentage of fat body mass  (OR 5.41; 95% CI 1.49-19.59; P = .01) were independent variables associated with vertebral fracture progression. The relationship between obesity and bone density is complex; higher estrogen levels derived from adipose tissue exert a protective effect on bone mineral density, whereas inflammatory cytokines and other hormones can adversely affect bone quality.[2] This study highlights the beneficial role of modalities such as diet and exercise to achieve healthy body mass, and how these can work in conjunction with antiresorptive therapy (such as denosumab) to benefit bone health in women receiving AI.

The phase 3 KEYNOTE-355 trial established the role of chemotherapy in combination with pembrolizumab in the first-line setting for programmed death-ligand 1 (PD-L1)–positive advanced triple-negative breast cancer (TNBC). Patients unselected for PD-L1 status in this trial who received platinum- or taxane-based chemotherapy with placebo had a median progression-free survival of 5.6 months.[3] Strategies to improve upon efficacy and tolerability are desired in this space, and various trials have evaluated "switch maintenance" that involves receipt of an intensive induction regimen followed by a switch to an alternative/more tolerable regimen after response is achieved.[4] The phase II DORA trial randomized 45 patients with advanced TNBC and ongoing stable disease or complete or partial response from first- or second-line platinum-based chemotherapy to a maintenance regimen of olaparib (300 mg orally twice daily) with or without durvalumab (1500 mg on day 1 and every 4 weeks) (Tan et al). At a median follow-up of 9.8 months, median progression-free survival was 4.0 months (95% CI 2.6-6.1) with olaparib and 6.1 months (95% CI 3.7-10.1) with the combination; both were significantly longer than the historical control of continued platinum-based therapy (P = .0023 and P < .0001, respectively). Durable disease control appeared more pronounced in patients with complete or partial response to prior platinum therapy, and no new safety signals were observed. Future efforts to study this approach include the phase 2/3 KEYLYNK-009 trial, which is evaluating olaparib plus pembrolizumab maintenance therapy after first-line chemotherapy plus pembrolizumab for TNBC.[5]

TNBC is a heterogenous subtype, characterized by aggressive biology, and it benefits from chemotherapy and immunotherapy treatment approaches. Presently, the management of early-stage TNBC often involves neoadjuvant systemic therapy; however, a proportion of patients receive treatment in the postoperative setting, highlighting the relevance of time to initiation of adjuvant therapy as well.[6] Various prior studies have showed that delayed administration of adjuvant chemotherapy for EBC can lead to adverse survival outcomes. Furthermore, this effect is subtype-dependent, with more aggressive tumors (luminal B, triple-negative, human epidermal growth factor receptor 2 [HER2]-positive) exhibiting inferior outcomes with delayed chemotherapy.[7] A retrospective cohort study that included 245 patients with early TNBC who received adjuvant chemotherapy after surgery evaluated the impact of time to initiation of adjuvant therapy in this population (Hatzipanagiotou et al). Superior survival outcomes were observed for the group receiving systemic therapy 22-28 days after surgery (median overall survival 10.2 years) compared with those receiving adjuvant chemotherapy at later time points (29-35 days, 36-42 days, and >6 weeks after surgery; median overall survival 8.3 years, 7.8 years, and 6.9 years, respectively). Patients receiving chemotherapy 22-28 days after surgery had significantly better survival than those receiving chemotherapy 29-35 days (P = .043) and >6 weeks (P = 0.033) postoperatively. This study emphasizes the importance of timely administration of adjuvant chemotherapy for early TNBC, and efforts aimed to identify potential challenges and propose solutions to optimize outcomes in this space are valuable.

Additional References

  1. Gnant M, Frantal S, Pfeiler G, et al, for the Austrian Breast & Colorectal Cancer Study Group. Long-term outcomes of adjuvant denosumab in breast cancer. NEJM Evid. 2022;1:EVIDoa2200162. doi: 10.1056/EVIDoa2200162 Source
  2. Fassio A, Idolazzi L, Rossini M, et al. The obesity paradox and osteoporosis. Eating and Weight Disorders - Studies on Anorexia, Bulimia and Obesity. 2018;23:293-30 doi: 10.1007/s40519-018-0505-2 Source
  3. Cortes J, Cescon DW, Rugo HS, et al, for the KEYNOTE-355 Investigators. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 2020;396:1817-1828. doi: 10.1016/S0140-6736(20)32531-9 Source
  4. Bachelot T, Filleron T, Bieche I, et al. Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: The randomized phase II SAFIR02-BREAST IMMUNO trial. Nat Med. 2021;27:250-255. doi: 10.1038/s41591-020-01189-2 Source
  5. Saji S, Cussac AL, Andre F, et al. 68TiP KEYLYNK-009: a phase II/III, open-label, randomized study of pembrolizumab (pembro) + olaparib (ola) vs pembro + chemotherapy after induction with first-line (1L) pembro + chemo in patients (pts) with locally recurrent inoperable or metastatic TNBC (abstract). Ann Oncol. 2020;31(Suppl 6):S1268. doi: 10.1016/j.annonc.2020.10.088 Source
  6. Ortmann O, Blohmer JU, Sibert NT, et al for 55 breast cancer centers certified by the German Cancer Society. Current clinical practice and outcome of neoadjuvant chemotherapy for early breast cancer: Analysis of individual data from 94,638 patients treated in 55 breast cancer centers. J Cancer Res Clin Oncol. 2023;149:1195-1209. doi: 10.1007/s00432-022-03938-x Source
  7. Yu KD, Fan L, Qiu LX, et al. Influence of delayed initiation of adjuvant chemotherapy on breast cancer survival is subtype-dependent. Oncotarget. 2017;8:46549-46556. doi: 10.18632/oncotarget.10551 Source
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 02/27/2024 - 13:30
Un-Gate On Date
Tue, 02/27/2024 - 13:30
Use ProPublica
CFC Schedule Remove Status
Tue, 02/27/2024 - 13:30
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
Activity Salesforce Deliverable ID
398326.1
Activity ID
109750
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Perjeta [ 3532 ]

Commentary: Medication Timing and Other Dupilumab Concerns, March 2024

Article Type
Changed
Wed, 03/13/2024 - 14:26
Dr. Feldman scans the journals, so you don’t have to!

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Ahn and colleagues found that having atopic dermatitis (AD) is associated with a higher risk for subsequent autoimmune disease. Is it a significant increased risk? What do we mean by "significant"? If we mean that the difference they observed wasn't likely due to chance alone, then yes, the observed difference was significant in the sense of being statistically significant. But what we really want to know is whether the difference they saw was large enough that someone with AD should lose sleep over it; I don't think they should. About six in 1000 patients per year without AD had a subsequent autoimmune disease; about eight in 1000 patients per year with AD had a subsequent autoimmune disease. There may be some genetic propensity to autoimmune disease in people with AD, but the great majority of people with AD, like the great majority of people in general, will not develop a subsequent autoimmune disease.

When skin diseases affect the palm or sole, they can have a disproportionately large negative effect on patients' lives. Hand and foot dermatitis can be disabling. Simpson and colleagues find that dupilumab is an effective treatment for AD of the hands and feet. Having safe and effective treatment for hand and foot dermatitis will be life-changing for many of our patients.

Patients often do very well with biologic treatment. When they do, they often wonder, Do I need to continue taking the medication? Lasheras-Pérez and colleagues found that the great majority of patients doing well taking dupilumab for AD could stretch out their dosing interval. I suspect a lot of our patients are doing this already. I used to worry that stretching out the dosing interval might lead to antidrug antibodies and loss of activity. Such loss of activity doesn't appear common. Because we also have multiple alternative treatments for severe AD, I think it may be quite reasonable for patients to try spreading out their doses after their disease has been well controlled for a good long time.

Superficial skin infections aren't rare in children, particularly children with AD. Paller and colleagues' study is informative about the safety of dupilumab in children. The drug, which blocks a pathway of the immune system, was associated with fewer infections. This is good news. The reduction in infections could be through restoring "immune balance" (whatever that means) or by improving skin barrier function. Perhaps the low rate of infection explains why dupilumab is not considered immunosuppressive.

I love studies of drug survival because I think that knowing the percentage of patients who stay with drug treatment is a good measure of overall safety and efficacy. Pezzolo and colleagues found — perhaps not surprisingly given the extraordinary efficacy of upadacitinib for AD — that almost no one discontinued the drug over 1.5 years due to lack of efficacy. There were patients who discontinued due to adverse events (and additional patients lost to follow-up who perhaps also discontinued the drug), but 80% of patients were still in the study at the end of 1.5 years. Three patients who weren't vaccinated for shingles developed shingles; encouraging patients to get the shingles vaccine may be a prudent measure when starting patients taking upadacitinib. 
 

Author and Disclosure Information

Steven R. Feldman, MD, PhD
Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

Publications
Topics
Sections
Author and Disclosure Information

Steven R. Feldman, MD, PhD
Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

Author and Disclosure Information

Steven R. Feldman, MD, PhD
Professor of Dermatology, Pathology and Social Sciences & Health Policy Wake Forest University School of Medicine, Winston-Salem, NC

Dr. Feldman scans the journals, so you don’t have to!
Dr. Feldman scans the journals, so you don’t have to!

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Ahn and colleagues found that having atopic dermatitis (AD) is associated with a higher risk for subsequent autoimmune disease. Is it a significant increased risk? What do we mean by "significant"? If we mean that the difference they observed wasn't likely due to chance alone, then yes, the observed difference was significant in the sense of being statistically significant. But what we really want to know is whether the difference they saw was large enough that someone with AD should lose sleep over it; I don't think they should. About six in 1000 patients per year without AD had a subsequent autoimmune disease; about eight in 1000 patients per year with AD had a subsequent autoimmune disease. There may be some genetic propensity to autoimmune disease in people with AD, but the great majority of people with AD, like the great majority of people in general, will not develop a subsequent autoimmune disease.

When skin diseases affect the palm or sole, they can have a disproportionately large negative effect on patients' lives. Hand and foot dermatitis can be disabling. Simpson and colleagues find that dupilumab is an effective treatment for AD of the hands and feet. Having safe and effective treatment for hand and foot dermatitis will be life-changing for many of our patients.

Patients often do very well with biologic treatment. When they do, they often wonder, Do I need to continue taking the medication? Lasheras-Pérez and colleagues found that the great majority of patients doing well taking dupilumab for AD could stretch out their dosing interval. I suspect a lot of our patients are doing this already. I used to worry that stretching out the dosing interval might lead to antidrug antibodies and loss of activity. Such loss of activity doesn't appear common. Because we also have multiple alternative treatments for severe AD, I think it may be quite reasonable for patients to try spreading out their doses after their disease has been well controlled for a good long time.

Superficial skin infections aren't rare in children, particularly children with AD. Paller and colleagues' study is informative about the safety of dupilumab in children. The drug, which blocks a pathway of the immune system, was associated with fewer infections. This is good news. The reduction in infections could be through restoring "immune balance" (whatever that means) or by improving skin barrier function. Perhaps the low rate of infection explains why dupilumab is not considered immunosuppressive.

I love studies of drug survival because I think that knowing the percentage of patients who stay with drug treatment is a good measure of overall safety and efficacy. Pezzolo and colleagues found — perhaps not surprisingly given the extraordinary efficacy of upadacitinib for AD — that almost no one discontinued the drug over 1.5 years due to lack of efficacy. There were patients who discontinued due to adverse events (and additional patients lost to follow-up who perhaps also discontinued the drug), but 80% of patients were still in the study at the end of 1.5 years. Three patients who weren't vaccinated for shingles developed shingles; encouraging patients to get the shingles vaccine may be a prudent measure when starting patients taking upadacitinib. 
 

feldman.steven.jpg
%3Cp%3E%3Cspan%20style%3D%22font-size%3A12.0pt%22%3E%3Cspan%20style%3D%22background%3Awhite%22%3E%3Cspan%20style%3D%22line-height%3A107%25%22%3E%3Cspan%20style%3D%22font-family%3A%26quot%3BArial%26quot%3B%2Csans-serif%22%3E%3Cspan%20style%3D%22color%3Ablack%22%3ESteven%20R.%20Feldman%2C%20MD%2C%20PhD%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fspan%3E%3C%2Fp%3E
Ahn and colleagues found that having atopic dermatitis (AD) is associated with a higher risk for subsequent autoimmune disease. Is it a significant increased risk? What do we mean by "significant"? If we mean that the difference they observed wasn't likely due to chance alone, then yes, the observed difference was significant in the sense of being statistically significant. But what we really want to know is whether the difference they saw was large enough that someone with AD should lose sleep over it; I don't think they should. About six in 1000 patients per year without AD had a subsequent autoimmune disease; about eight in 1000 patients per year with AD had a subsequent autoimmune disease. There may be some genetic propensity to autoimmune disease in people with AD, but the great majority of people with AD, like the great majority of people in general, will not develop a subsequent autoimmune disease.

When skin diseases affect the palm or sole, they can have a disproportionately large negative effect on patients' lives. Hand and foot dermatitis can be disabling. Simpson and colleagues find that dupilumab is an effective treatment for AD of the hands and feet. Having safe and effective treatment for hand and foot dermatitis will be life-changing for many of our patients.

Patients often do very well with biologic treatment. When they do, they often wonder, Do I need to continue taking the medication? Lasheras-Pérez and colleagues found that the great majority of patients doing well taking dupilumab for AD could stretch out their dosing interval. I suspect a lot of our patients are doing this already. I used to worry that stretching out the dosing interval might lead to antidrug antibodies and loss of activity. Such loss of activity doesn't appear common. Because we also have multiple alternative treatments for severe AD, I think it may be quite reasonable for patients to try spreading out their doses after their disease has been well controlled for a good long time.

Superficial skin infections aren't rare in children, particularly children with AD. Paller and colleagues' study is informative about the safety of dupilumab in children. The drug, which blocks a pathway of the immune system, was associated with fewer infections. This is good news. The reduction in infections could be through restoring "immune balance" (whatever that means) or by improving skin barrier function. Perhaps the low rate of infection explains why dupilumab is not considered immunosuppressive.

I love studies of drug survival because I think that knowing the percentage of patients who stay with drug treatment is a good measure of overall safety and efficacy. Pezzolo and colleagues found — perhaps not surprisingly given the extraordinary efficacy of upadacitinib for AD — that almost no one discontinued the drug over 1.5 years due to lack of efficacy. There were patients who discontinued due to adverse events (and additional patients lost to follow-up who perhaps also discontinued the drug), but 80% of patients were still in the study at the end of 1.5 years. Three patients who weren't vaccinated for shingles developed shingles; encouraging patients to get the shingles vaccine may be a prudent measure when starting patients taking upadacitinib. 
 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 02/27/2024 - 13:15
Un-Gate On Date
Tue, 02/27/2024 - 13:15
Use ProPublica
CFC Schedule Remove Status
Tue, 02/27/2024 - 13:15
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
Activity Salesforce Deliverable ID
400680.1
Activity ID
110501
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
RINVOQ [ 5260 ]

Commentary: Allergies, EDN, and the Psychosocial Burden of EoE, February 2024

Article Type
Changed
Mon, 04/22/2024 - 13:22
Dr Puerta scans the journals so you don't have to!

dr_puerta.jpg
%3Cp%3ECristian%20Puerta%2C%20M.D.%3C%2Fp%3E

A significant gap in our understanding of eosinophilic esophagitis (EoE) lies in how environmental factors, such as allergens or food, influence the response to proton pump inhibitor (PPI) therapy. While PPI achieve histologic remission in approximately 50% of patients, the response in the remaining 50% remains unclear. Addressing this, Muftah and colleagues conducted a study to evaluate the relationship between environmental and food allergies and PPI response in newly diagnosed EoE patients.

 

Between 2012 and 2016, adult patients newly diagnosed with EoE were tested for environmental and food allergies. Following diagnosis, patients participated in an 8-week trial of twice-daily PPI therapy. The treatment's effectiveness was assessed through repeated upper endoscopies with esophageal biopsies.

 

The study's primary outcome was the histologic remission of EoE, defined as a decrease in eosinophils to < 15 eosinophils/high-powered field (eos/hpf) in all esophageal biopsy samples during repeat endoscopy. Out of 61 patients, 21 achieved histologic remission, while 40 were classified as having PPI-nonresponding EoE. Among PPI-nonresponding EoE patients, positive food allergen testing was significantly more prevalent compared with PPI-responding EoE patients (82.5% vs 42.9%; P = .0003). Additionally, patients with >10 positive environmental allergen tests were significantly less likely to be PPI-responding EoE patients than those with <10 positive results (21% vs 53.9%; P = .03). A similar trend was observed in patients with >5 positive environmental allergens.

 

This study is not without limitations. It may exhibit a selection bias toward more severe cases and has a relatively small sample size, affecting its statistical power and generalizability.

 

This research supports the idea of more tailored management for EoE patients, focusing on their allergen profile, potentially leading to more effective treatment strategies and reducing unnecessary PPI trials. The statistically significant results pave the way for further research, providing an additional tool to predict PPI responsiveness and prevent delays in achieving remission.

 

Clinicians should consider patient characteristics, particularly positive food allergen tests, that might affect treatment response. More studies are needed, however, to understand the effect of environmental allergies on PPI response fully. A notable finding is that specific aeroallergens, such as oak, birch, Hormodendrum mold, dust mite (Dermatophagoides pteronyssinus), tree mix, and grass mix allergens, are associated with a lack of PPI response. This raises questions about whether exposure to these allergens during peak seasons could worsen PPI response in allergic EoE patients.

 

Key takeaways from this study include: (1) the importance of integrating allergen testing in EoE patients, especially those unresponsive to standard PPI therapy or suspected as having allergic phenotypes; (2) the need to monitor and adjust therapy based on clinical and histologic responses; and (3) the necessity of staying abreast of emerging research in this area.

 

EoE diagnosis presents unique challenges, particularly when patients exhibit exclusive distal esophageal eosinophilia or when discrepancies arise between endoscopic and histologic findings. Eosinophil-derived neurotoxin (EDN), a molecule previously studied for its role in monitoring allergy-mediated inflammatory diseases such as asthma and eczema, can shed light on these diagnostic difficulties.

 

Thomas and coworkers conducted a retrospective study in which they reviewed 231 pediatric patients, obtaining a minimum of four biopsies from at least two different levels of the esophagus. The study aimed to evaluate whether EDN concentrations, determined through esophageal epithelial brushing at the time of biopsy, could serve as an adjunctive diagnostic tool for EoE.

 

EDN levels proved sensitive (84.4%) and specific (94.6%) in evaluating active EoE when several measures of EoE were used in patients with active EoE compared with those with inactive EoE and the control group. Previous studies at the same institution had found EDN useful for differentiating EoE patients from non-EoE patients. Moreover, an EDN concentration > 10 μg/mL, when collected through esophageal epithelial brushing, was highly sensitive (97%) and specific (89%) for active EoE. This finding suggests the potential for using EDN as a biochemical marker, enhancing diagnostic accuracy and reducing the need for additional interventions in complex cases.

 

EDN as a biomarker could be invaluable for distinguishing difficult cases, such as those involving distal eosinophilia, active vs nonactive EoE, or non-EoE conditions, such as gastroesophageal reflux disease. Of note, lower EDN levels were observed in pediatric EoE patients who responded to PPI, suggesting EDN's potential utility in predicting PPI responsiveness. Incorporating the measurement of eosinophilic activity could add a new dimension to existing criteria, equipping clinicians with more precise diagnostic tools and reducing the reliance on multiple procedures. This approach would strengthen the correlation between symptomatic, endoscopic, and histologic data.

 

The study by Jensen and colleagues sheds light on a crucial aspect of EoE management: the psychosocial burden. A recent EoE diagnosis can be associated with increased symptom burden, somatization, and anxiety in patients and families, underscoring the need for a multidisciplinary approach to patient care that considers both physical and mental health. To date, numerous studies have focused on understanding the disease, its follow-up, and treatment. However, there has been limited exploration of the psychosocial burden and patient-associated factors in EoE.

 

In this context, this team aimed to enhance our understanding of the burden of EoE by evaluating psychosocial comorbidities, such as disordered sleep, anxiety, and somatization, in a pediatric population with EoE. The study included 87 patients of age 8-18 years who completed validated assessments during routine clinic visits, encompassing EoE symptoms (Pediatric Eosinophilic Esophagitis Symptom Scores, PEESSv2.0), quality of life (PedsQL-EoE), anxiety state and trait (State-Trait Anxiety Inventory for Children, STAI-C), somatization (Children's Somatic Symptoms Inventory-24, CSSI-24), and sleep-disordered breathing (University of Michigan Pediatric Sleep Questionnaire, PSQ).

 

The mean age of the participants was 12.8 years, highlighting the importance of addressing psychosocial distress in this age group, which undergoes crucial developmental stages. Most patients (82%, 71) had been diagnosed with EoE at least 12 months prior, and 60% (52) were treated with multiple approaches. Additionally, 34% (29) had undergone seven or more esophagogastroduodenoscopies, and nearly one third (33%, 27) had experienced a gastrointestinal-related emergency department visit. These factors potentially increase patient stress due to the continuous need for repeat procedures and hospital visits. An intriguing finding was that patients with shorter disease durations (6-12 months since diagnosis) experienced higher symptom burdens (P = .03). Patients with public insurance had less favorable scores for sleep-disordered breathing (P = .01).

 

Significantly, patients with neurodevelopmental comorbidities had higher scores for somatic symptoms, trait anxiety, and sleep-disordered breathing, and lower quality-of-life scores, compared with those without such comorbidities (P < .01 for all), suggesting that patients with neurodevelopmental issues might particularly benefit from tailored treatments addressing these aspects of the disease. Furthermore, patients with shorter disease durations since diagnosis exhibited higher somatic symptoms and trait anxiety (both P < .01). The study also revealed that patients with fewer esophagogastroduodenoscopies (1-3) had higher somatic symptom scores (P < .01), state anxiety (P = .02), and trait anxiety (P = .03). EoE-associated symptom burden was significantly correlated with increased somatic symptoms (0.34; 95% CI 0.23-0.45) and decreased quality of life (-0.42; 95% CI -0.59 to -0.25). Concerns about eating food and EoE-associated symptoms were both linked to the EoE-associated symptom burden.

 

This study has several limitations, including a relatively small sample size, which decreases the power and limits inferences for smaller groups within the sample. There was also an imbalance in gender distribution, with only 26% of patients being female, potentially limiting the generalizability of the findings. Moreover, the study included only EoE patients, lacking a control group for comparison to the general pediatric population.

 

Highlighting a significant aspect of pediatric EoE treatment, this study illuminates an area that might affect patients' long-term quality of life. It underscores the need for multidisciplinary care for EoE patients, where mental health professionals, such as psychologists or psychiatrists, can play a vital role in improving mental health through early identification and intervention for anxiety and somatization disorders. They can also provide education for patients and families on coping strategies. Peer support groups for children and adolescents could be another beneficial tool, allowing them to share experiences and reduce feelings of isolation.

 

Physicians who treat chronic diseases such as EoE should consider psychosocial factors, as they can affect both physical and mental quality of life. Using screening tools (such as PEESSv2.0, PedsQL-EoE, STAI-C, CSSI-24, or PSQ) during clinic visits can facilitate a more comprehensive evaluation.

Author and Disclosure Information

Research Fellow / Lab Manager

Thistlethwaite Lab BSB 4025

Division of Cardiothoracic Surgery

University of California, San Diego

Publications
Topics
Sections
Author and Disclosure Information

Research Fellow / Lab Manager

Thistlethwaite Lab BSB 4025

Division of Cardiothoracic Surgery

University of California, San Diego

Author and Disclosure Information

Research Fellow / Lab Manager

Thistlethwaite Lab BSB 4025

Division of Cardiothoracic Surgery

University of California, San Diego

Dr Puerta scans the journals so you don't have to!
Dr Puerta scans the journals so you don't have to!

dr_puerta.jpg
%3Cp%3ECristian%20Puerta%2C%20M.D.%3C%2Fp%3E

A significant gap in our understanding of eosinophilic esophagitis (EoE) lies in how environmental factors, such as allergens or food, influence the response to proton pump inhibitor (PPI) therapy. While PPI achieve histologic remission in approximately 50% of patients, the response in the remaining 50% remains unclear. Addressing this, Muftah and colleagues conducted a study to evaluate the relationship between environmental and food allergies and PPI response in newly diagnosed EoE patients.

 

Between 2012 and 2016, adult patients newly diagnosed with EoE were tested for environmental and food allergies. Following diagnosis, patients participated in an 8-week trial of twice-daily PPI therapy. The treatment's effectiveness was assessed through repeated upper endoscopies with esophageal biopsies.

 

The study's primary outcome was the histologic remission of EoE, defined as a decrease in eosinophils to < 15 eosinophils/high-powered field (eos/hpf) in all esophageal biopsy samples during repeat endoscopy. Out of 61 patients, 21 achieved histologic remission, while 40 were classified as having PPI-nonresponding EoE. Among PPI-nonresponding EoE patients, positive food allergen testing was significantly more prevalent compared with PPI-responding EoE patients (82.5% vs 42.9%; P = .0003). Additionally, patients with >10 positive environmental allergen tests were significantly less likely to be PPI-responding EoE patients than those with <10 positive results (21% vs 53.9%; P = .03). A similar trend was observed in patients with >5 positive environmental allergens.

 

This study is not without limitations. It may exhibit a selection bias toward more severe cases and has a relatively small sample size, affecting its statistical power and generalizability.

 

This research supports the idea of more tailored management for EoE patients, focusing on their allergen profile, potentially leading to more effective treatment strategies and reducing unnecessary PPI trials. The statistically significant results pave the way for further research, providing an additional tool to predict PPI responsiveness and prevent delays in achieving remission.

 

Clinicians should consider patient characteristics, particularly positive food allergen tests, that might affect treatment response. More studies are needed, however, to understand the effect of environmental allergies on PPI response fully. A notable finding is that specific aeroallergens, such as oak, birch, Hormodendrum mold, dust mite (Dermatophagoides pteronyssinus), tree mix, and grass mix allergens, are associated with a lack of PPI response. This raises questions about whether exposure to these allergens during peak seasons could worsen PPI response in allergic EoE patients.

 

Key takeaways from this study include: (1) the importance of integrating allergen testing in EoE patients, especially those unresponsive to standard PPI therapy or suspected as having allergic phenotypes; (2) the need to monitor and adjust therapy based on clinical and histologic responses; and (3) the necessity of staying abreast of emerging research in this area.

 

EoE diagnosis presents unique challenges, particularly when patients exhibit exclusive distal esophageal eosinophilia or when discrepancies arise between endoscopic and histologic findings. Eosinophil-derived neurotoxin (EDN), a molecule previously studied for its role in monitoring allergy-mediated inflammatory diseases such as asthma and eczema, can shed light on these diagnostic difficulties.

 

Thomas and coworkers conducted a retrospective study in which they reviewed 231 pediatric patients, obtaining a minimum of four biopsies from at least two different levels of the esophagus. The study aimed to evaluate whether EDN concentrations, determined through esophageal epithelial brushing at the time of biopsy, could serve as an adjunctive diagnostic tool for EoE.

 

EDN levels proved sensitive (84.4%) and specific (94.6%) in evaluating active EoE when several measures of EoE were used in patients with active EoE compared with those with inactive EoE and the control group. Previous studies at the same institution had found EDN useful for differentiating EoE patients from non-EoE patients. Moreover, an EDN concentration > 10 μg/mL, when collected through esophageal epithelial brushing, was highly sensitive (97%) and specific (89%) for active EoE. This finding suggests the potential for using EDN as a biochemical marker, enhancing diagnostic accuracy and reducing the need for additional interventions in complex cases.

 

EDN as a biomarker could be invaluable for distinguishing difficult cases, such as those involving distal eosinophilia, active vs nonactive EoE, or non-EoE conditions, such as gastroesophageal reflux disease. Of note, lower EDN levels were observed in pediatric EoE patients who responded to PPI, suggesting EDN's potential utility in predicting PPI responsiveness. Incorporating the measurement of eosinophilic activity could add a new dimension to existing criteria, equipping clinicians with more precise diagnostic tools and reducing the reliance on multiple procedures. This approach would strengthen the correlation between symptomatic, endoscopic, and histologic data.

 

The study by Jensen and colleagues sheds light on a crucial aspect of EoE management: the psychosocial burden. A recent EoE diagnosis can be associated with increased symptom burden, somatization, and anxiety in patients and families, underscoring the need for a multidisciplinary approach to patient care that considers both physical and mental health. To date, numerous studies have focused on understanding the disease, its follow-up, and treatment. However, there has been limited exploration of the psychosocial burden and patient-associated factors in EoE.

 

In this context, this team aimed to enhance our understanding of the burden of EoE by evaluating psychosocial comorbidities, such as disordered sleep, anxiety, and somatization, in a pediatric population with EoE. The study included 87 patients of age 8-18 years who completed validated assessments during routine clinic visits, encompassing EoE symptoms (Pediatric Eosinophilic Esophagitis Symptom Scores, PEESSv2.0), quality of life (PedsQL-EoE), anxiety state and trait (State-Trait Anxiety Inventory for Children, STAI-C), somatization (Children's Somatic Symptoms Inventory-24, CSSI-24), and sleep-disordered breathing (University of Michigan Pediatric Sleep Questionnaire, PSQ).

 

The mean age of the participants was 12.8 years, highlighting the importance of addressing psychosocial distress in this age group, which undergoes crucial developmental stages. Most patients (82%, 71) had been diagnosed with EoE at least 12 months prior, and 60% (52) were treated with multiple approaches. Additionally, 34% (29) had undergone seven or more esophagogastroduodenoscopies, and nearly one third (33%, 27) had experienced a gastrointestinal-related emergency department visit. These factors potentially increase patient stress due to the continuous need for repeat procedures and hospital visits. An intriguing finding was that patients with shorter disease durations (6-12 months since diagnosis) experienced higher symptom burdens (P = .03). Patients with public insurance had less favorable scores for sleep-disordered breathing (P = .01).

 

Significantly, patients with neurodevelopmental comorbidities had higher scores for somatic symptoms, trait anxiety, and sleep-disordered breathing, and lower quality-of-life scores, compared with those without such comorbidities (P < .01 for all), suggesting that patients with neurodevelopmental issues might particularly benefit from tailored treatments addressing these aspects of the disease. Furthermore, patients with shorter disease durations since diagnosis exhibited higher somatic symptoms and trait anxiety (both P < .01). The study also revealed that patients with fewer esophagogastroduodenoscopies (1-3) had higher somatic symptom scores (P < .01), state anxiety (P = .02), and trait anxiety (P = .03). EoE-associated symptom burden was significantly correlated with increased somatic symptoms (0.34; 95% CI 0.23-0.45) and decreased quality of life (-0.42; 95% CI -0.59 to -0.25). Concerns about eating food and EoE-associated symptoms were both linked to the EoE-associated symptom burden.

 

This study has several limitations, including a relatively small sample size, which decreases the power and limits inferences for smaller groups within the sample. There was also an imbalance in gender distribution, with only 26% of patients being female, potentially limiting the generalizability of the findings. Moreover, the study included only EoE patients, lacking a control group for comparison to the general pediatric population.

 

Highlighting a significant aspect of pediatric EoE treatment, this study illuminates an area that might affect patients' long-term quality of life. It underscores the need for multidisciplinary care for EoE patients, where mental health professionals, such as psychologists or psychiatrists, can play a vital role in improving mental health through early identification and intervention for anxiety and somatization disorders. They can also provide education for patients and families on coping strategies. Peer support groups for children and adolescents could be another beneficial tool, allowing them to share experiences and reduce feelings of isolation.

 

Physicians who treat chronic diseases such as EoE should consider psychosocial factors, as they can affect both physical and mental quality of life. Using screening tools (such as PEESSv2.0, PedsQL-EoE, STAI-C, CSSI-24, or PSQ) during clinic visits can facilitate a more comprehensive evaluation.

dr_puerta.jpg
%3Cp%3ECristian%20Puerta%2C%20M.D.%3C%2Fp%3E

A significant gap in our understanding of eosinophilic esophagitis (EoE) lies in how environmental factors, such as allergens or food, influence the response to proton pump inhibitor (PPI) therapy. While PPI achieve histologic remission in approximately 50% of patients, the response in the remaining 50% remains unclear. Addressing this, Muftah and colleagues conducted a study to evaluate the relationship between environmental and food allergies and PPI response in newly diagnosed EoE patients.

 

Between 2012 and 2016, adult patients newly diagnosed with EoE were tested for environmental and food allergies. Following diagnosis, patients participated in an 8-week trial of twice-daily PPI therapy. The treatment's effectiveness was assessed through repeated upper endoscopies with esophageal biopsies.

 

The study's primary outcome was the histologic remission of EoE, defined as a decrease in eosinophils to < 15 eosinophils/high-powered field (eos/hpf) in all esophageal biopsy samples during repeat endoscopy. Out of 61 patients, 21 achieved histologic remission, while 40 were classified as having PPI-nonresponding EoE. Among PPI-nonresponding EoE patients, positive food allergen testing was significantly more prevalent compared with PPI-responding EoE patients (82.5% vs 42.9%; P = .0003). Additionally, patients with >10 positive environmental allergen tests were significantly less likely to be PPI-responding EoE patients than those with <10 positive results (21% vs 53.9%; P = .03). A similar trend was observed in patients with >5 positive environmental allergens.

 

This study is not without limitations. It may exhibit a selection bias toward more severe cases and has a relatively small sample size, affecting its statistical power and generalizability.

 

This research supports the idea of more tailored management for EoE patients, focusing on their allergen profile, potentially leading to more effective treatment strategies and reducing unnecessary PPI trials. The statistically significant results pave the way for further research, providing an additional tool to predict PPI responsiveness and prevent delays in achieving remission.

 

Clinicians should consider patient characteristics, particularly positive food allergen tests, that might affect treatment response. More studies are needed, however, to understand the effect of environmental allergies on PPI response fully. A notable finding is that specific aeroallergens, such as oak, birch, Hormodendrum mold, dust mite (Dermatophagoides pteronyssinus), tree mix, and grass mix allergens, are associated with a lack of PPI response. This raises questions about whether exposure to these allergens during peak seasons could worsen PPI response in allergic EoE patients.

 

Key takeaways from this study include: (1) the importance of integrating allergen testing in EoE patients, especially those unresponsive to standard PPI therapy or suspected as having allergic phenotypes; (2) the need to monitor and adjust therapy based on clinical and histologic responses; and (3) the necessity of staying abreast of emerging research in this area.

 

EoE diagnosis presents unique challenges, particularly when patients exhibit exclusive distal esophageal eosinophilia or when discrepancies arise between endoscopic and histologic findings. Eosinophil-derived neurotoxin (EDN), a molecule previously studied for its role in monitoring allergy-mediated inflammatory diseases such as asthma and eczema, can shed light on these diagnostic difficulties.

 

Thomas and coworkers conducted a retrospective study in which they reviewed 231 pediatric patients, obtaining a minimum of four biopsies from at least two different levels of the esophagus. The study aimed to evaluate whether EDN concentrations, determined through esophageal epithelial brushing at the time of biopsy, could serve as an adjunctive diagnostic tool for EoE.

 

EDN levels proved sensitive (84.4%) and specific (94.6%) in evaluating active EoE when several measures of EoE were used in patients with active EoE compared with those with inactive EoE and the control group. Previous studies at the same institution had found EDN useful for differentiating EoE patients from non-EoE patients. Moreover, an EDN concentration > 10 μg/mL, when collected through esophageal epithelial brushing, was highly sensitive (97%) and specific (89%) for active EoE. This finding suggests the potential for using EDN as a biochemical marker, enhancing diagnostic accuracy and reducing the need for additional interventions in complex cases.

 

EDN as a biomarker could be invaluable for distinguishing difficult cases, such as those involving distal eosinophilia, active vs nonactive EoE, or non-EoE conditions, such as gastroesophageal reflux disease. Of note, lower EDN levels were observed in pediatric EoE patients who responded to PPI, suggesting EDN's potential utility in predicting PPI responsiveness. Incorporating the measurement of eosinophilic activity could add a new dimension to existing criteria, equipping clinicians with more precise diagnostic tools and reducing the reliance on multiple procedures. This approach would strengthen the correlation between symptomatic, endoscopic, and histologic data.

 

The study by Jensen and colleagues sheds light on a crucial aspect of EoE management: the psychosocial burden. A recent EoE diagnosis can be associated with increased symptom burden, somatization, and anxiety in patients and families, underscoring the need for a multidisciplinary approach to patient care that considers both physical and mental health. To date, numerous studies have focused on understanding the disease, its follow-up, and treatment. However, there has been limited exploration of the psychosocial burden and patient-associated factors in EoE.

 

In this context, this team aimed to enhance our understanding of the burden of EoE by evaluating psychosocial comorbidities, such as disordered sleep, anxiety, and somatization, in a pediatric population with EoE. The study included 87 patients of age 8-18 years who completed validated assessments during routine clinic visits, encompassing EoE symptoms (Pediatric Eosinophilic Esophagitis Symptom Scores, PEESSv2.0), quality of life (PedsQL-EoE), anxiety state and trait (State-Trait Anxiety Inventory for Children, STAI-C), somatization (Children's Somatic Symptoms Inventory-24, CSSI-24), and sleep-disordered breathing (University of Michigan Pediatric Sleep Questionnaire, PSQ).

 

The mean age of the participants was 12.8 years, highlighting the importance of addressing psychosocial distress in this age group, which undergoes crucial developmental stages. Most patients (82%, 71) had been diagnosed with EoE at least 12 months prior, and 60% (52) were treated with multiple approaches. Additionally, 34% (29) had undergone seven or more esophagogastroduodenoscopies, and nearly one third (33%, 27) had experienced a gastrointestinal-related emergency department visit. These factors potentially increase patient stress due to the continuous need for repeat procedures and hospital visits. An intriguing finding was that patients with shorter disease durations (6-12 months since diagnosis) experienced higher symptom burdens (P = .03). Patients with public insurance had less favorable scores for sleep-disordered breathing (P = .01).

 

Significantly, patients with neurodevelopmental comorbidities had higher scores for somatic symptoms, trait anxiety, and sleep-disordered breathing, and lower quality-of-life scores, compared with those without such comorbidities (P < .01 for all), suggesting that patients with neurodevelopmental issues might particularly benefit from tailored treatments addressing these aspects of the disease. Furthermore, patients with shorter disease durations since diagnosis exhibited higher somatic symptoms and trait anxiety (both P < .01). The study also revealed that patients with fewer esophagogastroduodenoscopies (1-3) had higher somatic symptom scores (P < .01), state anxiety (P = .02), and trait anxiety (P = .03). EoE-associated symptom burden was significantly correlated with increased somatic symptoms (0.34; 95% CI 0.23-0.45) and decreased quality of life (-0.42; 95% CI -0.59 to -0.25). Concerns about eating food and EoE-associated symptoms were both linked to the EoE-associated symptom burden.

 

This study has several limitations, including a relatively small sample size, which decreases the power and limits inferences for smaller groups within the sample. There was also an imbalance in gender distribution, with only 26% of patients being female, potentially limiting the generalizability of the findings. Moreover, the study included only EoE patients, lacking a control group for comparison to the general pediatric population.

 

Highlighting a significant aspect of pediatric EoE treatment, this study illuminates an area that might affect patients' long-term quality of life. It underscores the need for multidisciplinary care for EoE patients, where mental health professionals, such as psychologists or psychiatrists, can play a vital role in improving mental health through early identification and intervention for anxiety and somatization disorders. They can also provide education for patients and families on coping strategies. Peer support groups for children and adolescents could be another beneficial tool, allowing them to share experiences and reduce feelings of isolation.

 

Physicians who treat chronic diseases such as EoE should consider psychosocial factors, as they can affect both physical and mental quality of life. Using screening tools (such as PEESSv2.0, PedsQL-EoE, STAI-C, CSSI-24, or PSQ) during clinic visits can facilitate a more comprehensive evaluation.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Fri, 12/29/2023 - 17:15
Un-Gate On Date
Fri, 12/29/2023 - 17:15
Use ProPublica
CFC Schedule Remove Status
Fri, 12/29/2023 - 17:15
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