Strategies to Improve Long-Term Outcomes in Younger Patients With Hodgkin Lymphoma

Article Type
Changed
Mon, 06/26/2023 - 12:27
Display Headline
Strategies to Improve Long-Term Outcomes in Younger Patients With Hodgkin Lymphoma

Ann LaCasce, MD, MMSc
The current treatments for classical Hodgkin lymphoma (cHL) in adolescents and young adults (AYA) are associated with high rates of remission but may lead to treatment-related complications years later. These problems, such as organ damage and secondary malignancies that arise long after otherwise effective treatment, are a threat to long-term outcomes. This threat is seen especially in the AYA population because of their longer life expectancy. Concerns such as cardiovascular effects and second cancers in the AYA population are paramount, emphasizing the importance of identifying safer regimens for these individuals. Initiatives to incorporate risk-adapted treatment regimens and novel therapies with a lower risk of late-occurring complications are being actively pursued. This review highlights the potential of several of these initiatives for AYA patients.


Background

Hodgkin lymphoma occurs in fewer than 9,000 individuals in the United States each year,1 but it is one of the most common types of cancer in AYAs.2 For the purposes of cHL, AYA is typically defined as an age range of 18 to 39 years, which covers the first of 2 bimodal peaks in incidence but stops short of the second.3,4 The first of these peaks occurs between the ages of 15 and 34 years, while the second begins at about age 55.Children younger than 15 years of age can also develop Hodgkin lymphoma, but it is less common.6

In AYAs and in adults, more than 90% of patients with Hodgkin lymphoma have cHL.7 Most AYAs present with the nodular sclerosis subtype, but cHL is managed differently in pediatric patients versus in adult centers.8,9 Evidence suggests that the specific risks of common treatment protocols, although similar, are not the same in AYAs as in adults.10,11 Even though the literature evaluating the presentation and management of AYA cHL has been growing since 2005, when the AYA Oncology Progress Review Group called for AYAs to be recognized
as a distinct group, clinical trials specific to AYA cHL remain limited.9

Major Hodgkin lymphoma guidelines only partially address AYAs as a distinct group. In guidelines issued by the National Cancer Institute, the differences in clinical presentation of AYAs are described for young children, AYAs, and older adults, but there are no treatment recommendations specific to AYAs.12 Guidelines from the EuroNet Paediatric Hodgkin Lymphoma Group offer recommendations for relapsed and refractory Hodgkin lymphoma, but do not differentiate between children and adolescents.13 The National Comprehensive Cancer Network (NCCN) provides separate treatment recommendations for patients 18 years or younger and those who are older than 18.14,15 For Hodgkin lymphoma, AYA is not addressed as a separate category even though the NCCN has provided general guidelines for treatment of malignancies in AYA.16

First-line therapies are effective in children, AYAs, and adults. Survival rates at 5 years have increased steadily, approaching or exceeding 90% across age groups even for patients with unfavorable risk characteristics.17 This success has permitted greater focus on developing strategies that preserve efficacy with lower acute and long-term risks.

Risk-Adapted Therapies

While the potential for new and novel therapies to reduce the risk of long-term toxicities continues to be explored, adjusting existing regimens to reduce these risks has proven to be a viable strategy. This adjustment is a standard of care in the pediatric setting based on results from such studies as German GPOH-HD 95, which suggested that doses of radiotherapy, a major contributor to late toxicities,18 can be omitted in patients with a complete response after chemotherapy.11 This pediatric trial contained both younger children and adolescents, but subsequent secondary analyses looking specifically at AYAs in this and other trials have suggested that efficacy is similarly preserved with risk-adapted strategies.9

However, due to AYA patients with cHL being treated using both pediatric and adult approaches, the persistent debate about optimal therapies in this age group complicates the effort to define a well-accepted strategy for risk adjustment. While risk-adapted strategies that rely on interim positron emission tomography (PET) to calibrate treatment intensity are now being used routinely across age stratifications, other initiatives are creating additional opportunities to gauge the impact on late effects in AYAs. These include strategies to improve collaboration across groups of trialists and data generated by observational cohorts, which can evaluate late effects not captured in time-limited clinical trials.

Among recent data supporting risk-adjusted therapy, the toxicity outcomes from a multicenter trial of PET-guided intensive treatment in patients with newly diagnosed advanced cHL were presented at the 2022 annual meeting of the American Society of Hematology.19 This phase 3 trial enrolled patients younger than 60 years, 79% of whom were younger than 45 years. Building on previous evidence that PET guidance improves the safety of eBEACOPP (escalated doses of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone), nearly 1,500 patients were randomized to this strategy or to PET-guided BrECADD, a modified eBEACOPP in which the antibody conjugate brentuximab vedotin (BV) was substituted for bleomycin. For an adjudicated endpoint of treatment-related morbidity, the experimental BrECAAD regimen reduced the risk by nearly 30% (hazard ratio [HR] 0.72). It is unclear whether this strategy will be used in the United States, where trials have been built on ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) rather than BEACOPP.

Efficacy data from this trial are not yet available, and these data will be important. There is concern that PET-directed therapy might result in lower toxicity at a cost of reduced rates of disease control. It is possible that the serious consequences of late toxicities—including infertility, compromised cardiovascular function, secondary cancers, and other organ damage—might need to be balanced against some loss of efficacy.

Novel Targeted Therapies

The goal of reducing late toxicities of cHL therapy in AYAs is also likely to be advanced by novel therapies. Research endeavors include a multicenter collaboration between US and Canadian investigators that is exploring the combination of nivolumab (a checkpoint inhibitor) plus BV.20 The trial recently completed accrual and includes both adult and pediatric patients. If novel agents prove effective for improving efficacy while reducing the risk of late complications in AYAs, they are expected to have a profound effect on clinical practice.

Arguably, the era of targeted and novel therapies in cHL was initiated more than 10 years ago with the introduction of BV for the treatment of advanced disease in older adults.21 BV was moved into the front line for patients 18 years of age or older with advanced cHL in a trial that compared the standard of ABVD to the same drugs with BV substituted for bleomycin.22 In this study, the BV-containing regimen was associated with a significantly improved progression-free survival (PFS) (P = .04) and a lower rate of adverse events, including pulmonary toxicity (1% vs 3%) after 2 years of follow-up.

A similar study recently associated a BV-containing regimen with even greater efficacy in pediatric high-risk cHL.23 In this multicenter study with 600 treatment-naïve patients ranging in age from 2 to 21 years, the standard pediatric regimen of doxorubicin, bleomycin, vincristine, etoposide, prednisone, and cyclophosphamide was compared to the same regimen with BV substituted for bleomycin. With event-free survival as the primary endpoint, the experimental regimen was associated with a nearly 60% reduction in the risk of an adverse event or death (HR 0.41). However, no substantial differences were noted in toxicity after a follow-up of 42 months. It not yet clear whether the elimination of bleomycin will translate into less late toxicity, such as pulmonary or cardiovascular morbidity.

In the era of targeted therapies, the experience with BV has been a step toward more effective treatments using novel mechanisms of action to improve outcomes when used in the first-line treatment of patients with high-risk disease. Historically, many regimens and treatments that have demonstrated efficacy in relapsed and refractory cHL have found their way into the first-line setting. This trend might also be true of the checkpoint inhibitors, which have been tested extensively in relapsed/refractory cHL. In AYA patients with cHL, the rationale for these treatments might not only include a poor predicted response to current regimens, but a reduced risk of late toxicities if long-term follow-up demonstrates these treatments reduce late complications, such as secondary malignancies, which are associated with standard strategies, particularly those that include radiotherapy.

If targeted therapies do preserve efficacy and reduce risk of late complications, strategies to individualize therapy will remain relevant. Many of the emerging targeted therapies involve challenging and costly treatment protocols that demand selective application. Efforts to develop simpler and more precise biomarkers might streamline this task. Of promising developments in this area, cell-free DNA (cfDNA) appears to be near routine clinical application. A small study of cfDNA conducted in 121 patients found that minimal residual disease assessment by repeat cfDNA sequencing predicted response and PFS when performed as early as a week after treatment initiation.24 If larger studies confirm accuracy, this biomarker strategy might prove simpler and more convenient than PET imaging.

Summary

In the treatment of hematologic malignancies, cHL is widely regarded as a success story with high rates of extended survival among children, AYAs, and older adults. This level of success does not obviate the need for even more effective treatments, and also permits more attention to be directed to reducing the risk of late toxicities. For the AYA population, which represents a large group with cHL, the current directions of clinical research offer the promise of imminent changes in how the disease is controlled and a reduction in treatment-related late morbidity and mortality.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Hodgkin Lymphoma. American Cancer Society. Accessed March 20, 2023. https://www.cancer.org/cancer/hodgkin-lymphoma.html
  2. Aben KK, van Gaal C, van Gils NA, van der Graaf WT, Zielhuis GA. Cancer in adolescents and young adults (15-29 years): a population-based study in the Netherlands 1989-2009. Acta Oncol. 2012;51(7):922-933. doi:10.3109/0284186X.2012.705891
  3. Ansell SM. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and
    management. Am J Hematol. 2016;91(4):434-442. doi:10.1002/ajh.24272
  4. Cartwright RA, Watkins G. Epidemiology of Hodgkin’s disease: a review. Hematol Oncol. 2004;22(1):11-26. doi:10.1002/hon.723
  5. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N Engl J Med. 1998;339(21):1506-1514. doi:10.1056/NEJM199811193392104
  6. Bleyer A, Barr R, Hayes-Lattin B, et al. The distinctive biology of cancer in adolescents and young adults. Nat Rev Cancer. 2008;8(4):288-298. doi:10.1038/nrc2349
  7. Shanbhag S, Ambinder RF. Hodgkin lymphoma: a review and update on recent progress. CA Cancer J Clin. 2018;68(2):116-132. doi:10.3322/caac.21438
  8. Bigenwald C, Galimard JE, Quero L, et al. Hodgkin lymphoma in adolescent and young adults: insights from an adult tertiary single-center cohort of 349 patients. Oncotarget. 2017;8(45):80073-80082. doi:10.18632/oncotarget.20684
  9. Kahn JM, Kelly KM. Adolescent and young adult Hodgkin lymphoma: raising the bar through collaborative science and multidisciplinary care. Pediatr Blood Cancer. 2018;65(7):e27033. doi:10.1002/pbc.27033
  10. Yung L, Smith P, Hancock BW, et al. Long term outcome in adolescents with Hodgkin’s lymphoma: poor results using regimens designed for adults. Leuk Lymphoma. 2004;45(8):1579-1585. doi:10.1080/1042819042000209404
  11. Dorffel W, Ruhl U, Luders H, et al. Treatment of children and adolescents with Hodgkin lymphoma without radiotherapy for patients in complete remission after chemotherapy: final results of the multinational trial GPOH-HD95. J Clin Oncol. 2013;31(12):1562-1568. doi:10.1200/JCO.2012.45.3266
  12. National Cancer Institute. Childhood Hodgkin lymphoma treatment (PDQ®)–Health Professional Version. National Institutes of Health. Updated February 14, 2023. Accessed March 20, 2023. https://www.cancer.gov/types/lymphoma/hp/child-hodgkin-treatment-pdq
  13. Daw S, Hasenclever D, Mascarin M, et al. Risk and response adapted treatment guidelines for managing first relapsed and refractory classical Hodgkin lymphoma in children and young people. Recommendations from the EuroNet Pediatric Hodgkin Lymphoma Group. Hemasphere. 2020;4(1):e329. doi:10.1097/HS9.0000000000000329
  14. Flerlage JE, Hiniker SM, Armenian S, et al. Pediatric Hodgkin lymphoma, version 3.2021. J Natl Compr Canc Netw. 2021;19(6):733-754. doi:10.6004/jnccn.2021.0027
  15. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Hodgkin lymphoma. Version 2.2023. November 8, 2022. Accessed March 20, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hodgkins.pdf
  16. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Adolescent and young adult (AYA) oncology. Version 3.2023. January 9, 2023. Accessed March 20, 2023. https://www.nccn.org/professionals/physician_gls/pdf/aya.pdf
  17. Mohty R, Dulery R, Bazarbachi AH, et al. Latest advances in the management of classical Hodgkin lymphoma: the era of novel therapies. Blood Cancer J. 2021;11(7):126. doi:10.1038/s41408-021-00518-z
  18. Witkowska M, Majchrzak A, Smolewski P. The role of radiotherapy in Hodgkin’s lymphoma: what has been achieved during the last 50 years? Biomed Res Int. 2015;2015:485071. doi:10.1155/2015/485071
  19. Borchmann P, Moccia A, Greil R, et al. Treatment-related morbidity in patients with classical Hodgkin lymphoma: results of the ongoing, randomized phase II HD21 trial by the German Hodgkin Study Group. Hemasphere. 2022;6(suppl ):1-2. doi:10.1097/01.HS9.0000890576.23258.1c
  20. Immunotherapy (nivolumab or brentuximab vedotin) plus combination chemotherapy in treating patients with newly diagnosed stage III-IV classic Hodgkin lymphoma. ClinicalTrials.gov. Updated March 8, 2023. Accessed March 20, 2023. https://clinicaltrials.gov/ct2/show/NCT03907488
  21. Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol. 2012;30(18):2183-2189. doi:10.1200/JCO.2011.38.0410
  22. Connors JM, Jurczak W, Straus DJ, et al. Brentuximab vedotin with chemotherapy for stage III or IV Hodgkin’s lymphoma. N Engl J Med. 2018;378(4):331-344. doi:10.1056/NEJMoa1708984
  23. Castellino SM, Pei Q, Parsons SK, et al. Brentuximab vedotin with chemotherapy in pediatric high-risk Hodgkin’s lymphoma. N Engl J Med. 2022;387(18):1649-1660. doi:10.1056/NEJMoa2206660
  24. Sobesky S, Mammadova L, Cirillo M, et al. In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. Med (N Y). 2021;2(10):1171-1193.e11. doi:10.1016/j.medj.2021.09.002
Author and Disclosure Information

Ann LaCasce, MD, MMSc
Associate Professor, Hematology and Medical Oncology
Dana Farber Cancer Center
Program Director, Dana Farber MGB Fellowship in Hematology/Oncology
Harvard Medical School
Boston, MA

Ann S. LaCasce, MD, MMSc, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for Kite Pharma; Seagen Inc.

Serve(d) as a speaker or a member of a speakers bureau for Research to Practice®.

Received income in an amount equal to or greater than $250 from Kite Pharma; Seagen Inc.

Publications
Topics
Author and Disclosure Information

Ann LaCasce, MD, MMSc
Associate Professor, Hematology and Medical Oncology
Dana Farber Cancer Center
Program Director, Dana Farber MGB Fellowship in Hematology/Oncology
Harvard Medical School
Boston, MA

Ann S. LaCasce, MD, MMSc, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for Kite Pharma; Seagen Inc.

Serve(d) as a speaker or a member of a speakers bureau for Research to Practice®.

Received income in an amount equal to or greater than $250 from Kite Pharma; Seagen Inc.

Author and Disclosure Information

Ann LaCasce, MD, MMSc
Associate Professor, Hematology and Medical Oncology
Dana Farber Cancer Center
Program Director, Dana Farber MGB Fellowship in Hematology/Oncology
Harvard Medical School
Boston, MA

Ann S. LaCasce, MD, MMSc, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for Kite Pharma; Seagen Inc.

Serve(d) as a speaker or a member of a speakers bureau for Research to Practice®.

Received income in an amount equal to or greater than $250 from Kite Pharma; Seagen Inc.

Ann LaCasce, MD, MMSc
The current treatments for classical Hodgkin lymphoma (cHL) in adolescents and young adults (AYA) are associated with high rates of remission but may lead to treatment-related complications years later. These problems, such as organ damage and secondary malignancies that arise long after otherwise effective treatment, are a threat to long-term outcomes. This threat is seen especially in the AYA population because of their longer life expectancy. Concerns such as cardiovascular effects and second cancers in the AYA population are paramount, emphasizing the importance of identifying safer regimens for these individuals. Initiatives to incorporate risk-adapted treatment regimens and novel therapies with a lower risk of late-occurring complications are being actively pursued. This review highlights the potential of several of these initiatives for AYA patients.


Background

Hodgkin lymphoma occurs in fewer than 9,000 individuals in the United States each year,1 but it is one of the most common types of cancer in AYAs.2 For the purposes of cHL, AYA is typically defined as an age range of 18 to 39 years, which covers the first of 2 bimodal peaks in incidence but stops short of the second.3,4 The first of these peaks occurs between the ages of 15 and 34 years, while the second begins at about age 55.Children younger than 15 years of age can also develop Hodgkin lymphoma, but it is less common.6

In AYAs and in adults, more than 90% of patients with Hodgkin lymphoma have cHL.7 Most AYAs present with the nodular sclerosis subtype, but cHL is managed differently in pediatric patients versus in adult centers.8,9 Evidence suggests that the specific risks of common treatment protocols, although similar, are not the same in AYAs as in adults.10,11 Even though the literature evaluating the presentation and management of AYA cHL has been growing since 2005, when the AYA Oncology Progress Review Group called for AYAs to be recognized
as a distinct group, clinical trials specific to AYA cHL remain limited.9

Major Hodgkin lymphoma guidelines only partially address AYAs as a distinct group. In guidelines issued by the National Cancer Institute, the differences in clinical presentation of AYAs are described for young children, AYAs, and older adults, but there are no treatment recommendations specific to AYAs.12 Guidelines from the EuroNet Paediatric Hodgkin Lymphoma Group offer recommendations for relapsed and refractory Hodgkin lymphoma, but do not differentiate between children and adolescents.13 The National Comprehensive Cancer Network (NCCN) provides separate treatment recommendations for patients 18 years or younger and those who are older than 18.14,15 For Hodgkin lymphoma, AYA is not addressed as a separate category even though the NCCN has provided general guidelines for treatment of malignancies in AYA.16

First-line therapies are effective in children, AYAs, and adults. Survival rates at 5 years have increased steadily, approaching or exceeding 90% across age groups even for patients with unfavorable risk characteristics.17 This success has permitted greater focus on developing strategies that preserve efficacy with lower acute and long-term risks.

Risk-Adapted Therapies

While the potential for new and novel therapies to reduce the risk of long-term toxicities continues to be explored, adjusting existing regimens to reduce these risks has proven to be a viable strategy. This adjustment is a standard of care in the pediatric setting based on results from such studies as German GPOH-HD 95, which suggested that doses of radiotherapy, a major contributor to late toxicities,18 can be omitted in patients with a complete response after chemotherapy.11 This pediatric trial contained both younger children and adolescents, but subsequent secondary analyses looking specifically at AYAs in this and other trials have suggested that efficacy is similarly preserved with risk-adapted strategies.9

However, due to AYA patients with cHL being treated using both pediatric and adult approaches, the persistent debate about optimal therapies in this age group complicates the effort to define a well-accepted strategy for risk adjustment. While risk-adapted strategies that rely on interim positron emission tomography (PET) to calibrate treatment intensity are now being used routinely across age stratifications, other initiatives are creating additional opportunities to gauge the impact on late effects in AYAs. These include strategies to improve collaboration across groups of trialists and data generated by observational cohorts, which can evaluate late effects not captured in time-limited clinical trials.

Among recent data supporting risk-adjusted therapy, the toxicity outcomes from a multicenter trial of PET-guided intensive treatment in patients with newly diagnosed advanced cHL were presented at the 2022 annual meeting of the American Society of Hematology.19 This phase 3 trial enrolled patients younger than 60 years, 79% of whom were younger than 45 years. Building on previous evidence that PET guidance improves the safety of eBEACOPP (escalated doses of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone), nearly 1,500 patients were randomized to this strategy or to PET-guided BrECADD, a modified eBEACOPP in which the antibody conjugate brentuximab vedotin (BV) was substituted for bleomycin. For an adjudicated endpoint of treatment-related morbidity, the experimental BrECAAD regimen reduced the risk by nearly 30% (hazard ratio [HR] 0.72). It is unclear whether this strategy will be used in the United States, where trials have been built on ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) rather than BEACOPP.

Efficacy data from this trial are not yet available, and these data will be important. There is concern that PET-directed therapy might result in lower toxicity at a cost of reduced rates of disease control. It is possible that the serious consequences of late toxicities—including infertility, compromised cardiovascular function, secondary cancers, and other organ damage—might need to be balanced against some loss of efficacy.

Novel Targeted Therapies

The goal of reducing late toxicities of cHL therapy in AYAs is also likely to be advanced by novel therapies. Research endeavors include a multicenter collaboration between US and Canadian investigators that is exploring the combination of nivolumab (a checkpoint inhibitor) plus BV.20 The trial recently completed accrual and includes both adult and pediatric patients. If novel agents prove effective for improving efficacy while reducing the risk of late complications in AYAs, they are expected to have a profound effect on clinical practice.

Arguably, the era of targeted and novel therapies in cHL was initiated more than 10 years ago with the introduction of BV for the treatment of advanced disease in older adults.21 BV was moved into the front line for patients 18 years of age or older with advanced cHL in a trial that compared the standard of ABVD to the same drugs with BV substituted for bleomycin.22 In this study, the BV-containing regimen was associated with a significantly improved progression-free survival (PFS) (P = .04) and a lower rate of adverse events, including pulmonary toxicity (1% vs 3%) after 2 years of follow-up.

A similar study recently associated a BV-containing regimen with even greater efficacy in pediatric high-risk cHL.23 In this multicenter study with 600 treatment-naïve patients ranging in age from 2 to 21 years, the standard pediatric regimen of doxorubicin, bleomycin, vincristine, etoposide, prednisone, and cyclophosphamide was compared to the same regimen with BV substituted for bleomycin. With event-free survival as the primary endpoint, the experimental regimen was associated with a nearly 60% reduction in the risk of an adverse event or death (HR 0.41). However, no substantial differences were noted in toxicity after a follow-up of 42 months. It not yet clear whether the elimination of bleomycin will translate into less late toxicity, such as pulmonary or cardiovascular morbidity.

In the era of targeted therapies, the experience with BV has been a step toward more effective treatments using novel mechanisms of action to improve outcomes when used in the first-line treatment of patients with high-risk disease. Historically, many regimens and treatments that have demonstrated efficacy in relapsed and refractory cHL have found their way into the first-line setting. This trend might also be true of the checkpoint inhibitors, which have been tested extensively in relapsed/refractory cHL. In AYA patients with cHL, the rationale for these treatments might not only include a poor predicted response to current regimens, but a reduced risk of late toxicities if long-term follow-up demonstrates these treatments reduce late complications, such as secondary malignancies, which are associated with standard strategies, particularly those that include radiotherapy.

If targeted therapies do preserve efficacy and reduce risk of late complications, strategies to individualize therapy will remain relevant. Many of the emerging targeted therapies involve challenging and costly treatment protocols that demand selective application. Efforts to develop simpler and more precise biomarkers might streamline this task. Of promising developments in this area, cell-free DNA (cfDNA) appears to be near routine clinical application. A small study of cfDNA conducted in 121 patients found that minimal residual disease assessment by repeat cfDNA sequencing predicted response and PFS when performed as early as a week after treatment initiation.24 If larger studies confirm accuracy, this biomarker strategy might prove simpler and more convenient than PET imaging.

Summary

In the treatment of hematologic malignancies, cHL is widely regarded as a success story with high rates of extended survival among children, AYAs, and older adults. This level of success does not obviate the need for even more effective treatments, and also permits more attention to be directed to reducing the risk of late toxicities. For the AYA population, which represents a large group with cHL, the current directions of clinical research offer the promise of imminent changes in how the disease is controlled and a reduction in treatment-related late morbidity and mortality.

Click to read more from 2023 Rare Diseases Report: Cancers

Ann LaCasce, MD, MMSc
The current treatments for classical Hodgkin lymphoma (cHL) in adolescents and young adults (AYA) are associated with high rates of remission but may lead to treatment-related complications years later. These problems, such as organ damage and secondary malignancies that arise long after otherwise effective treatment, are a threat to long-term outcomes. This threat is seen especially in the AYA population because of their longer life expectancy. Concerns such as cardiovascular effects and second cancers in the AYA population are paramount, emphasizing the importance of identifying safer regimens for these individuals. Initiatives to incorporate risk-adapted treatment regimens and novel therapies with a lower risk of late-occurring complications are being actively pursued. This review highlights the potential of several of these initiatives for AYA patients.


Background

Hodgkin lymphoma occurs in fewer than 9,000 individuals in the United States each year,1 but it is one of the most common types of cancer in AYAs.2 For the purposes of cHL, AYA is typically defined as an age range of 18 to 39 years, which covers the first of 2 bimodal peaks in incidence but stops short of the second.3,4 The first of these peaks occurs between the ages of 15 and 34 years, while the second begins at about age 55.Children younger than 15 years of age can also develop Hodgkin lymphoma, but it is less common.6

In AYAs and in adults, more than 90% of patients with Hodgkin lymphoma have cHL.7 Most AYAs present with the nodular sclerosis subtype, but cHL is managed differently in pediatric patients versus in adult centers.8,9 Evidence suggests that the specific risks of common treatment protocols, although similar, are not the same in AYAs as in adults.10,11 Even though the literature evaluating the presentation and management of AYA cHL has been growing since 2005, when the AYA Oncology Progress Review Group called for AYAs to be recognized
as a distinct group, clinical trials specific to AYA cHL remain limited.9

Major Hodgkin lymphoma guidelines only partially address AYAs as a distinct group. In guidelines issued by the National Cancer Institute, the differences in clinical presentation of AYAs are described for young children, AYAs, and older adults, but there are no treatment recommendations specific to AYAs.12 Guidelines from the EuroNet Paediatric Hodgkin Lymphoma Group offer recommendations for relapsed and refractory Hodgkin lymphoma, but do not differentiate between children and adolescents.13 The National Comprehensive Cancer Network (NCCN) provides separate treatment recommendations for patients 18 years or younger and those who are older than 18.14,15 For Hodgkin lymphoma, AYA is not addressed as a separate category even though the NCCN has provided general guidelines for treatment of malignancies in AYA.16

First-line therapies are effective in children, AYAs, and adults. Survival rates at 5 years have increased steadily, approaching or exceeding 90% across age groups even for patients with unfavorable risk characteristics.17 This success has permitted greater focus on developing strategies that preserve efficacy with lower acute and long-term risks.

Risk-Adapted Therapies

While the potential for new and novel therapies to reduce the risk of long-term toxicities continues to be explored, adjusting existing regimens to reduce these risks has proven to be a viable strategy. This adjustment is a standard of care in the pediatric setting based on results from such studies as German GPOH-HD 95, which suggested that doses of radiotherapy, a major contributor to late toxicities,18 can be omitted in patients with a complete response after chemotherapy.11 This pediatric trial contained both younger children and adolescents, but subsequent secondary analyses looking specifically at AYAs in this and other trials have suggested that efficacy is similarly preserved with risk-adapted strategies.9

However, due to AYA patients with cHL being treated using both pediatric and adult approaches, the persistent debate about optimal therapies in this age group complicates the effort to define a well-accepted strategy for risk adjustment. While risk-adapted strategies that rely on interim positron emission tomography (PET) to calibrate treatment intensity are now being used routinely across age stratifications, other initiatives are creating additional opportunities to gauge the impact on late effects in AYAs. These include strategies to improve collaboration across groups of trialists and data generated by observational cohorts, which can evaluate late effects not captured in time-limited clinical trials.

Among recent data supporting risk-adjusted therapy, the toxicity outcomes from a multicenter trial of PET-guided intensive treatment in patients with newly diagnosed advanced cHL were presented at the 2022 annual meeting of the American Society of Hematology.19 This phase 3 trial enrolled patients younger than 60 years, 79% of whom were younger than 45 years. Building on previous evidence that PET guidance improves the safety of eBEACOPP (escalated doses of bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone), nearly 1,500 patients were randomized to this strategy or to PET-guided BrECADD, a modified eBEACOPP in which the antibody conjugate brentuximab vedotin (BV) was substituted for bleomycin. For an adjudicated endpoint of treatment-related morbidity, the experimental BrECAAD regimen reduced the risk by nearly 30% (hazard ratio [HR] 0.72). It is unclear whether this strategy will be used in the United States, where trials have been built on ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) rather than BEACOPP.

Efficacy data from this trial are not yet available, and these data will be important. There is concern that PET-directed therapy might result in lower toxicity at a cost of reduced rates of disease control. It is possible that the serious consequences of late toxicities—including infertility, compromised cardiovascular function, secondary cancers, and other organ damage—might need to be balanced against some loss of efficacy.

Novel Targeted Therapies

The goal of reducing late toxicities of cHL therapy in AYAs is also likely to be advanced by novel therapies. Research endeavors include a multicenter collaboration between US and Canadian investigators that is exploring the combination of nivolumab (a checkpoint inhibitor) plus BV.20 The trial recently completed accrual and includes both adult and pediatric patients. If novel agents prove effective for improving efficacy while reducing the risk of late complications in AYAs, they are expected to have a profound effect on clinical practice.

Arguably, the era of targeted and novel therapies in cHL was initiated more than 10 years ago with the introduction of BV for the treatment of advanced disease in older adults.21 BV was moved into the front line for patients 18 years of age or older with advanced cHL in a trial that compared the standard of ABVD to the same drugs with BV substituted for bleomycin.22 In this study, the BV-containing regimen was associated with a significantly improved progression-free survival (PFS) (P = .04) and a lower rate of adverse events, including pulmonary toxicity (1% vs 3%) after 2 years of follow-up.

A similar study recently associated a BV-containing regimen with even greater efficacy in pediatric high-risk cHL.23 In this multicenter study with 600 treatment-naïve patients ranging in age from 2 to 21 years, the standard pediatric regimen of doxorubicin, bleomycin, vincristine, etoposide, prednisone, and cyclophosphamide was compared to the same regimen with BV substituted for bleomycin. With event-free survival as the primary endpoint, the experimental regimen was associated with a nearly 60% reduction in the risk of an adverse event or death (HR 0.41). However, no substantial differences were noted in toxicity after a follow-up of 42 months. It not yet clear whether the elimination of bleomycin will translate into less late toxicity, such as pulmonary or cardiovascular morbidity.

In the era of targeted therapies, the experience with BV has been a step toward more effective treatments using novel mechanisms of action to improve outcomes when used in the first-line treatment of patients with high-risk disease. Historically, many regimens and treatments that have demonstrated efficacy in relapsed and refractory cHL have found their way into the first-line setting. This trend might also be true of the checkpoint inhibitors, which have been tested extensively in relapsed/refractory cHL. In AYA patients with cHL, the rationale for these treatments might not only include a poor predicted response to current regimens, but a reduced risk of late toxicities if long-term follow-up demonstrates these treatments reduce late complications, such as secondary malignancies, which are associated with standard strategies, particularly those that include radiotherapy.

If targeted therapies do preserve efficacy and reduce risk of late complications, strategies to individualize therapy will remain relevant. Many of the emerging targeted therapies involve challenging and costly treatment protocols that demand selective application. Efforts to develop simpler and more precise biomarkers might streamline this task. Of promising developments in this area, cell-free DNA (cfDNA) appears to be near routine clinical application. A small study of cfDNA conducted in 121 patients found that minimal residual disease assessment by repeat cfDNA sequencing predicted response and PFS when performed as early as a week after treatment initiation.24 If larger studies confirm accuracy, this biomarker strategy might prove simpler and more convenient than PET imaging.

Summary

In the treatment of hematologic malignancies, cHL is widely regarded as a success story with high rates of extended survival among children, AYAs, and older adults. This level of success does not obviate the need for even more effective treatments, and also permits more attention to be directed to reducing the risk of late toxicities. For the AYA population, which represents a large group with cHL, the current directions of clinical research offer the promise of imminent changes in how the disease is controlled and a reduction in treatment-related late morbidity and mortality.

Click to read more from 2023 Rare Diseases Report: Cancers

References
  1. Hodgkin Lymphoma. American Cancer Society. Accessed March 20, 2023. https://www.cancer.org/cancer/hodgkin-lymphoma.html
  2. Aben KK, van Gaal C, van Gils NA, van der Graaf WT, Zielhuis GA. Cancer in adolescents and young adults (15-29 years): a population-based study in the Netherlands 1989-2009. Acta Oncol. 2012;51(7):922-933. doi:10.3109/0284186X.2012.705891
  3. Ansell SM. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and
    management. Am J Hematol. 2016;91(4):434-442. doi:10.1002/ajh.24272
  4. Cartwright RA, Watkins G. Epidemiology of Hodgkin’s disease: a review. Hematol Oncol. 2004;22(1):11-26. doi:10.1002/hon.723
  5. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N Engl J Med. 1998;339(21):1506-1514. doi:10.1056/NEJM199811193392104
  6. Bleyer A, Barr R, Hayes-Lattin B, et al. The distinctive biology of cancer in adolescents and young adults. Nat Rev Cancer. 2008;8(4):288-298. doi:10.1038/nrc2349
  7. Shanbhag S, Ambinder RF. Hodgkin lymphoma: a review and update on recent progress. CA Cancer J Clin. 2018;68(2):116-132. doi:10.3322/caac.21438
  8. Bigenwald C, Galimard JE, Quero L, et al. Hodgkin lymphoma in adolescent and young adults: insights from an adult tertiary single-center cohort of 349 patients. Oncotarget. 2017;8(45):80073-80082. doi:10.18632/oncotarget.20684
  9. Kahn JM, Kelly KM. Adolescent and young adult Hodgkin lymphoma: raising the bar through collaborative science and multidisciplinary care. Pediatr Blood Cancer. 2018;65(7):e27033. doi:10.1002/pbc.27033
  10. Yung L, Smith P, Hancock BW, et al. Long term outcome in adolescents with Hodgkin’s lymphoma: poor results using regimens designed for adults. Leuk Lymphoma. 2004;45(8):1579-1585. doi:10.1080/1042819042000209404
  11. Dorffel W, Ruhl U, Luders H, et al. Treatment of children and adolescents with Hodgkin lymphoma without radiotherapy for patients in complete remission after chemotherapy: final results of the multinational trial GPOH-HD95. J Clin Oncol. 2013;31(12):1562-1568. doi:10.1200/JCO.2012.45.3266
  12. National Cancer Institute. Childhood Hodgkin lymphoma treatment (PDQ®)–Health Professional Version. National Institutes of Health. Updated February 14, 2023. Accessed March 20, 2023. https://www.cancer.gov/types/lymphoma/hp/child-hodgkin-treatment-pdq
  13. Daw S, Hasenclever D, Mascarin M, et al. Risk and response adapted treatment guidelines for managing first relapsed and refractory classical Hodgkin lymphoma in children and young people. Recommendations from the EuroNet Pediatric Hodgkin Lymphoma Group. Hemasphere. 2020;4(1):e329. doi:10.1097/HS9.0000000000000329
  14. Flerlage JE, Hiniker SM, Armenian S, et al. Pediatric Hodgkin lymphoma, version 3.2021. J Natl Compr Canc Netw. 2021;19(6):733-754. doi:10.6004/jnccn.2021.0027
  15. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Hodgkin lymphoma. Version 2.2023. November 8, 2022. Accessed March 20, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hodgkins.pdf
  16. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Adolescent and young adult (AYA) oncology. Version 3.2023. January 9, 2023. Accessed March 20, 2023. https://www.nccn.org/professionals/physician_gls/pdf/aya.pdf
  17. Mohty R, Dulery R, Bazarbachi AH, et al. Latest advances in the management of classical Hodgkin lymphoma: the era of novel therapies. Blood Cancer J. 2021;11(7):126. doi:10.1038/s41408-021-00518-z
  18. Witkowska M, Majchrzak A, Smolewski P. The role of radiotherapy in Hodgkin’s lymphoma: what has been achieved during the last 50 years? Biomed Res Int. 2015;2015:485071. doi:10.1155/2015/485071
  19. Borchmann P, Moccia A, Greil R, et al. Treatment-related morbidity in patients with classical Hodgkin lymphoma: results of the ongoing, randomized phase II HD21 trial by the German Hodgkin Study Group. Hemasphere. 2022;6(suppl ):1-2. doi:10.1097/01.HS9.0000890576.23258.1c
  20. Immunotherapy (nivolumab or brentuximab vedotin) plus combination chemotherapy in treating patients with newly diagnosed stage III-IV classic Hodgkin lymphoma. ClinicalTrials.gov. Updated March 8, 2023. Accessed March 20, 2023. https://clinicaltrials.gov/ct2/show/NCT03907488
  21. Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol. 2012;30(18):2183-2189. doi:10.1200/JCO.2011.38.0410
  22. Connors JM, Jurczak W, Straus DJ, et al. Brentuximab vedotin with chemotherapy for stage III or IV Hodgkin’s lymphoma. N Engl J Med. 2018;378(4):331-344. doi:10.1056/NEJMoa1708984
  23. Castellino SM, Pei Q, Parsons SK, et al. Brentuximab vedotin with chemotherapy in pediatric high-risk Hodgkin’s lymphoma. N Engl J Med. 2022;387(18):1649-1660. doi:10.1056/NEJMoa2206660
  24. Sobesky S, Mammadova L, Cirillo M, et al. In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. Med (N Y). 2021;2(10):1171-1193.e11. doi:10.1016/j.medj.2021.09.002
References
  1. Hodgkin Lymphoma. American Cancer Society. Accessed March 20, 2023. https://www.cancer.org/cancer/hodgkin-lymphoma.html
  2. Aben KK, van Gaal C, van Gils NA, van der Graaf WT, Zielhuis GA. Cancer in adolescents and young adults (15-29 years): a population-based study in the Netherlands 1989-2009. Acta Oncol. 2012;51(7):922-933. doi:10.3109/0284186X.2012.705891
  3. Ansell SM. Hodgkin lymphoma: 2016 update on diagnosis, risk-stratification, and
    management. Am J Hematol. 2016;91(4):434-442. doi:10.1002/ajh.24272
  4. Cartwright RA, Watkins G. Epidemiology of Hodgkin’s disease: a review. Hematol Oncol. 2004;22(1):11-26. doi:10.1002/hon.723
  5. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N Engl J Med. 1998;339(21):1506-1514. doi:10.1056/NEJM199811193392104
  6. Bleyer A, Barr R, Hayes-Lattin B, et al. The distinctive biology of cancer in adolescents and young adults. Nat Rev Cancer. 2008;8(4):288-298. doi:10.1038/nrc2349
  7. Shanbhag S, Ambinder RF. Hodgkin lymphoma: a review and update on recent progress. CA Cancer J Clin. 2018;68(2):116-132. doi:10.3322/caac.21438
  8. Bigenwald C, Galimard JE, Quero L, et al. Hodgkin lymphoma in adolescent and young adults: insights from an adult tertiary single-center cohort of 349 patients. Oncotarget. 2017;8(45):80073-80082. doi:10.18632/oncotarget.20684
  9. Kahn JM, Kelly KM. Adolescent and young adult Hodgkin lymphoma: raising the bar through collaborative science and multidisciplinary care. Pediatr Blood Cancer. 2018;65(7):e27033. doi:10.1002/pbc.27033
  10. Yung L, Smith P, Hancock BW, et al. Long term outcome in adolescents with Hodgkin’s lymphoma: poor results using regimens designed for adults. Leuk Lymphoma. 2004;45(8):1579-1585. doi:10.1080/1042819042000209404
  11. Dorffel W, Ruhl U, Luders H, et al. Treatment of children and adolescents with Hodgkin lymphoma without radiotherapy for patients in complete remission after chemotherapy: final results of the multinational trial GPOH-HD95. J Clin Oncol. 2013;31(12):1562-1568. doi:10.1200/JCO.2012.45.3266
  12. National Cancer Institute. Childhood Hodgkin lymphoma treatment (PDQ®)–Health Professional Version. National Institutes of Health. Updated February 14, 2023. Accessed March 20, 2023. https://www.cancer.gov/types/lymphoma/hp/child-hodgkin-treatment-pdq
  13. Daw S, Hasenclever D, Mascarin M, et al. Risk and response adapted treatment guidelines for managing first relapsed and refractory classical Hodgkin lymphoma in children and young people. Recommendations from the EuroNet Pediatric Hodgkin Lymphoma Group. Hemasphere. 2020;4(1):e329. doi:10.1097/HS9.0000000000000329
  14. Flerlage JE, Hiniker SM, Armenian S, et al. Pediatric Hodgkin lymphoma, version 3.2021. J Natl Compr Canc Netw. 2021;19(6):733-754. doi:10.6004/jnccn.2021.0027
  15. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Hodgkin lymphoma. Version 2.2023. November 8, 2022. Accessed March 20, 2023. https://www.nccn.org/professionals/physician_gls/pdf/hodgkins.pdf
  16. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: Adolescent and young adult (AYA) oncology. Version 3.2023. January 9, 2023. Accessed March 20, 2023. https://www.nccn.org/professionals/physician_gls/pdf/aya.pdf
  17. Mohty R, Dulery R, Bazarbachi AH, et al. Latest advances in the management of classical Hodgkin lymphoma: the era of novel therapies. Blood Cancer J. 2021;11(7):126. doi:10.1038/s41408-021-00518-z
  18. Witkowska M, Majchrzak A, Smolewski P. The role of radiotherapy in Hodgkin’s lymphoma: what has been achieved during the last 50 years? Biomed Res Int. 2015;2015:485071. doi:10.1155/2015/485071
  19. Borchmann P, Moccia A, Greil R, et al. Treatment-related morbidity in patients with classical Hodgkin lymphoma: results of the ongoing, randomized phase II HD21 trial by the German Hodgkin Study Group. Hemasphere. 2022;6(suppl ):1-2. doi:10.1097/01.HS9.0000890576.23258.1c
  20. Immunotherapy (nivolumab or brentuximab vedotin) plus combination chemotherapy in treating patients with newly diagnosed stage III-IV classic Hodgkin lymphoma. ClinicalTrials.gov. Updated March 8, 2023. Accessed March 20, 2023. https://clinicaltrials.gov/ct2/show/NCT03907488
  21. Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol. 2012;30(18):2183-2189. doi:10.1200/JCO.2011.38.0410
  22. Connors JM, Jurczak W, Straus DJ, et al. Brentuximab vedotin with chemotherapy for stage III or IV Hodgkin’s lymphoma. N Engl J Med. 2018;378(4):331-344. doi:10.1056/NEJMoa1708984
  23. Castellino SM, Pei Q, Parsons SK, et al. Brentuximab vedotin with chemotherapy in pediatric high-risk Hodgkin’s lymphoma. N Engl J Med. 2022;387(18):1649-1660. doi:10.1056/NEJMoa2206660
  24. Sobesky S, Mammadova L, Cirillo M, et al. In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. Med (N Y). 2021;2(10):1171-1193.e11. doi:10.1016/j.medj.2021.09.002
Publications
Publications
Topics
Article Type
Display Headline
Strategies to Improve Long-Term Outcomes in Younger Patients With Hodgkin Lymphoma
Display Headline
Strategies to Improve Long-Term Outcomes in Younger Patients With Hodgkin Lymphoma
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 06/22/2023 - 08:45
Un-Gate On Date
Thu, 06/22/2023 - 08:45
Use ProPublica
CFC Schedule Remove Status
Thu, 06/22/2023 - 08:45
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

Patients With Newly Diagnosed Mantle Cell Lymphoma and the Relevance of Clinical Trials

Article Type
Changed
Wed, 05/03/2023 - 00:15

 

What is the significance of the recent TRIANGLE study on mantle cell lymphoma (MCL)?

Dr. LaCasce: The TRIANGLE study is extremely important in previously untreated, transplant-eligible patients with MCL. The cutoff age for transplants varies by center and is between 60 and 75 years. In the absence of a TP53 mutation, we have typically used induction chemotherapy followed by autologous stem-cell transplant (ASCT), followed by 3 years of maintenance rituximab. Obviously, this is a lot of therapy.

The TRIANGLE study was a 3-arm study in which ibrutinib-containing therapy was compared with standard RCHOP/RDHAP followed by ASCT. Maintenance rituximab became standard of care midway through the trial and was added. In the first experimental arm, ibrutinib was combined with RCHOP and then given as maintenance for 2 years following ASCT. The second experimental arm included the same schedule of ibrutinib and omitted the ASCT.

The results are early, but what has been presented thus far, ibrutinib induction and maintenance with ASCT is clearly superior to the standard arm with ASCT. Although the data are not statistically mature, the failure-free survival of the 2 ibrutinib arms was similar, suggesting that transplant may not be necessary. Longer follow-up is necessary to confirm this conclusion and assess overall survival in all 3 arms.

If the results hold, ASCT could become a thing of the past or perhaps used in the second line. With the favorable activity of chimeric antigen receptor (CAR) T-cell therapy, however, it is unclear whether ASCT would be used in second line. Avoiding the sequential use of ASCT and CAR T-cell therapy is appealing given the stem-cell damage that can result. It is appealing to think about not using ASCT upfront, because ASCT increases the risk of myelodysplastic syndrome.

The TRIANGLE data are likely to change the frontline management of MCL. Although ibrutinib was the first Bruton tyrosine kinase (BTK) inhibitor approved in MCL and has obviously changed the field dramatically, it is significantly less well-tolerated than the next generation of drugs—acalabrutinib and zanubrutinib. I suspect these will be substituted for ibrutinib and we will see even more tolerable upfront regimens for patients with newly diagnosed MCL.

Have there been any disparities that you found in patients newly diagnosed with MCL regarding age, sex, or ethnicity?

Dr. LaCasce: MCL typically affects patients in their 60s. It is rare in young patients, and approximately 75% of the cases are male. If you look at the demographics, it is more common in White patients and less common in Hispanic and African American patients. In addition, there is an association with farming, which likely contribute to the demographics of patients with MCL.

What is your recommended approach to managing patients newly diagnosed with MCL in your day-to-day practice?

Dr. LaCasce: Management is a bit tricky right now because the TRIANGLE study is not part of any guidelines thus far. Therefore, most would argue the standard treatment continues to include ASCT upfront. There is an important, large randomized  study (NCT03267433) going on in the United States that is assessing the role of ASCT in patients who are in MRD-negative complete remission at the end of induction therapy. These patients are randomized to ASCT plus maintenance rituximab versus maintenance rituximab alone.

We are still enrolling patients to participate in this study, which is addressing a different question than TRIANGLE. I think we will learn a lot from this study. For patients who are not interested in participating in this study, we talk about the risks and benefits of ASCT.

One or 2 years ago, I would have strongly encouraged patients who were appropriate candidates to consider transplant in first remission. With the TRIANGLE data, however, and now that we have CAR T-cell therapy, I think it is more important to tailor the recommendation to the individual patient. If a patient is reluctant about ASCT and the associated risks, I do not push it.

If patients want the most aggressive approach associated with the longest remissions, at this moment, before TRIANGLE findings have been adopted into guidelines, I continue to recommend ASCT. For patients who have TP53 mutation, however, we treat with typically less aggressive therapy, as this patient population does not benefit from ASCT. We look forward to more data incorporating BTK inhibitors upfront, particularly for this group of patients, who tend to have a more adverse prognosis.

Do you feel MCL data and clinical trials are important areas of focus for your colleagues?

Dr. LaCasce: Yes. I think it is a rapidly evolving field, which is really exciting. We are seeing data now from the bispecific antibodies in the relapsed/refractory setting. We also need more data using pirtobrutinib for patients who have had BTK inhibitors and compare pirtobrutinib (a non-covalent BTK inhibitor) with the covalent BTK inhibitors.

I would strongly encourage patients to participate in clinical trials so that we can better answer these important questions. When patients go online and read about MCL, they often see a median survival of 3 to 4 years, which is completely outdated. The overall prognosis of MCL has changed dramatically since I have been in the field. Hopefully, survival will continue to improve, and therapies will become more tolerable, as well.

Author and Disclosure Information

Ann S. LaCasce, MD, MMSc, Associate Professor, Department of Medical Oncology, Harvard Medical School; Program Director, Dana Farber MGB Fellowship in Hematology/Oncology, Institute Physician, Dana Farber Cancer Center, Boston, Massachusetts. 

Ann S. LaCasce, MD, MMSc, has disclosed the following relevant financial relationships: Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: Kite Pharma; Seagen. Serve(d) as a speaker or a member of a speaker’s bureau for: Research to Practice Inc. Received income in an amount equal to or greater than $250 from: Kite Pharma; Seagen.

Publications
Topics
Sections
Author and Disclosure Information

Ann S. LaCasce, MD, MMSc, Associate Professor, Department of Medical Oncology, Harvard Medical School; Program Director, Dana Farber MGB Fellowship in Hematology/Oncology, Institute Physician, Dana Farber Cancer Center, Boston, Massachusetts. 

Ann S. LaCasce, MD, MMSc, has disclosed the following relevant financial relationships: Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: Kite Pharma; Seagen. Serve(d) as a speaker or a member of a speaker’s bureau for: Research to Practice Inc. Received income in an amount equal to or greater than $250 from: Kite Pharma; Seagen.

Author and Disclosure Information

Ann S. LaCasce, MD, MMSc, Associate Professor, Department of Medical Oncology, Harvard Medical School; Program Director, Dana Farber MGB Fellowship in Hematology/Oncology, Institute Physician, Dana Farber Cancer Center, Boston, Massachusetts. 

Ann S. LaCasce, MD, MMSc, has disclosed the following relevant financial relationships: Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: Kite Pharma; Seagen. Serve(d) as a speaker or a member of a speaker’s bureau for: Research to Practice Inc. Received income in an amount equal to or greater than $250 from: Kite Pharma; Seagen.

 

What is the significance of the recent TRIANGLE study on mantle cell lymphoma (MCL)?

Dr. LaCasce: The TRIANGLE study is extremely important in previously untreated, transplant-eligible patients with MCL. The cutoff age for transplants varies by center and is between 60 and 75 years. In the absence of a TP53 mutation, we have typically used induction chemotherapy followed by autologous stem-cell transplant (ASCT), followed by 3 years of maintenance rituximab. Obviously, this is a lot of therapy.

The TRIANGLE study was a 3-arm study in which ibrutinib-containing therapy was compared with standard RCHOP/RDHAP followed by ASCT. Maintenance rituximab became standard of care midway through the trial and was added. In the first experimental arm, ibrutinib was combined with RCHOP and then given as maintenance for 2 years following ASCT. The second experimental arm included the same schedule of ibrutinib and omitted the ASCT.

The results are early, but what has been presented thus far, ibrutinib induction and maintenance with ASCT is clearly superior to the standard arm with ASCT. Although the data are not statistically mature, the failure-free survival of the 2 ibrutinib arms was similar, suggesting that transplant may not be necessary. Longer follow-up is necessary to confirm this conclusion and assess overall survival in all 3 arms.

If the results hold, ASCT could become a thing of the past or perhaps used in the second line. With the favorable activity of chimeric antigen receptor (CAR) T-cell therapy, however, it is unclear whether ASCT would be used in second line. Avoiding the sequential use of ASCT and CAR T-cell therapy is appealing given the stem-cell damage that can result. It is appealing to think about not using ASCT upfront, because ASCT increases the risk of myelodysplastic syndrome.

The TRIANGLE data are likely to change the frontline management of MCL. Although ibrutinib was the first Bruton tyrosine kinase (BTK) inhibitor approved in MCL and has obviously changed the field dramatically, it is significantly less well-tolerated than the next generation of drugs—acalabrutinib and zanubrutinib. I suspect these will be substituted for ibrutinib and we will see even more tolerable upfront regimens for patients with newly diagnosed MCL.

Have there been any disparities that you found in patients newly diagnosed with MCL regarding age, sex, or ethnicity?

Dr. LaCasce: MCL typically affects patients in their 60s. It is rare in young patients, and approximately 75% of the cases are male. If you look at the demographics, it is more common in White patients and less common in Hispanic and African American patients. In addition, there is an association with farming, which likely contribute to the demographics of patients with MCL.

What is your recommended approach to managing patients newly diagnosed with MCL in your day-to-day practice?

Dr. LaCasce: Management is a bit tricky right now because the TRIANGLE study is not part of any guidelines thus far. Therefore, most would argue the standard treatment continues to include ASCT upfront. There is an important, large randomized  study (NCT03267433) going on in the United States that is assessing the role of ASCT in patients who are in MRD-negative complete remission at the end of induction therapy. These patients are randomized to ASCT plus maintenance rituximab versus maintenance rituximab alone.

We are still enrolling patients to participate in this study, which is addressing a different question than TRIANGLE. I think we will learn a lot from this study. For patients who are not interested in participating in this study, we talk about the risks and benefits of ASCT.

One or 2 years ago, I would have strongly encouraged patients who were appropriate candidates to consider transplant in first remission. With the TRIANGLE data, however, and now that we have CAR T-cell therapy, I think it is more important to tailor the recommendation to the individual patient. If a patient is reluctant about ASCT and the associated risks, I do not push it.

If patients want the most aggressive approach associated with the longest remissions, at this moment, before TRIANGLE findings have been adopted into guidelines, I continue to recommend ASCT. For patients who have TP53 mutation, however, we treat with typically less aggressive therapy, as this patient population does not benefit from ASCT. We look forward to more data incorporating BTK inhibitors upfront, particularly for this group of patients, who tend to have a more adverse prognosis.

Do you feel MCL data and clinical trials are important areas of focus for your colleagues?

Dr. LaCasce: Yes. I think it is a rapidly evolving field, which is really exciting. We are seeing data now from the bispecific antibodies in the relapsed/refractory setting. We also need more data using pirtobrutinib for patients who have had BTK inhibitors and compare pirtobrutinib (a non-covalent BTK inhibitor) with the covalent BTK inhibitors.

I would strongly encourage patients to participate in clinical trials so that we can better answer these important questions. When patients go online and read about MCL, they often see a median survival of 3 to 4 years, which is completely outdated. The overall prognosis of MCL has changed dramatically since I have been in the field. Hopefully, survival will continue to improve, and therapies will become more tolerable, as well.

 

What is the significance of the recent TRIANGLE study on mantle cell lymphoma (MCL)?

Dr. LaCasce: The TRIANGLE study is extremely important in previously untreated, transplant-eligible patients with MCL. The cutoff age for transplants varies by center and is between 60 and 75 years. In the absence of a TP53 mutation, we have typically used induction chemotherapy followed by autologous stem-cell transplant (ASCT), followed by 3 years of maintenance rituximab. Obviously, this is a lot of therapy.

The TRIANGLE study was a 3-arm study in which ibrutinib-containing therapy was compared with standard RCHOP/RDHAP followed by ASCT. Maintenance rituximab became standard of care midway through the trial and was added. In the first experimental arm, ibrutinib was combined with RCHOP and then given as maintenance for 2 years following ASCT. The second experimental arm included the same schedule of ibrutinib and omitted the ASCT.

The results are early, but what has been presented thus far, ibrutinib induction and maintenance with ASCT is clearly superior to the standard arm with ASCT. Although the data are not statistically mature, the failure-free survival of the 2 ibrutinib arms was similar, suggesting that transplant may not be necessary. Longer follow-up is necessary to confirm this conclusion and assess overall survival in all 3 arms.

If the results hold, ASCT could become a thing of the past or perhaps used in the second line. With the favorable activity of chimeric antigen receptor (CAR) T-cell therapy, however, it is unclear whether ASCT would be used in second line. Avoiding the sequential use of ASCT and CAR T-cell therapy is appealing given the stem-cell damage that can result. It is appealing to think about not using ASCT upfront, because ASCT increases the risk of myelodysplastic syndrome.

The TRIANGLE data are likely to change the frontline management of MCL. Although ibrutinib was the first Bruton tyrosine kinase (BTK) inhibitor approved in MCL and has obviously changed the field dramatically, it is significantly less well-tolerated than the next generation of drugs—acalabrutinib and zanubrutinib. I suspect these will be substituted for ibrutinib and we will see even more tolerable upfront regimens for patients with newly diagnosed MCL.

Have there been any disparities that you found in patients newly diagnosed with MCL regarding age, sex, or ethnicity?

Dr. LaCasce: MCL typically affects patients in their 60s. It is rare in young patients, and approximately 75% of the cases are male. If you look at the demographics, it is more common in White patients and less common in Hispanic and African American patients. In addition, there is an association with farming, which likely contribute to the demographics of patients with MCL.

What is your recommended approach to managing patients newly diagnosed with MCL in your day-to-day practice?

Dr. LaCasce: Management is a bit tricky right now because the TRIANGLE study is not part of any guidelines thus far. Therefore, most would argue the standard treatment continues to include ASCT upfront. There is an important, large randomized  study (NCT03267433) going on in the United States that is assessing the role of ASCT in patients who are in MRD-negative complete remission at the end of induction therapy. These patients are randomized to ASCT plus maintenance rituximab versus maintenance rituximab alone.

We are still enrolling patients to participate in this study, which is addressing a different question than TRIANGLE. I think we will learn a lot from this study. For patients who are not interested in participating in this study, we talk about the risks and benefits of ASCT.

One or 2 years ago, I would have strongly encouraged patients who were appropriate candidates to consider transplant in first remission. With the TRIANGLE data, however, and now that we have CAR T-cell therapy, I think it is more important to tailor the recommendation to the individual patient. If a patient is reluctant about ASCT and the associated risks, I do not push it.

If patients want the most aggressive approach associated with the longest remissions, at this moment, before TRIANGLE findings have been adopted into guidelines, I continue to recommend ASCT. For patients who have TP53 mutation, however, we treat with typically less aggressive therapy, as this patient population does not benefit from ASCT. We look forward to more data incorporating BTK inhibitors upfront, particularly for this group of patients, who tend to have a more adverse prognosis.

Do you feel MCL data and clinical trials are important areas of focus for your colleagues?

Dr. LaCasce: Yes. I think it is a rapidly evolving field, which is really exciting. We are seeing data now from the bispecific antibodies in the relapsed/refractory setting. We also need more data using pirtobrutinib for patients who have had BTK inhibitors and compare pirtobrutinib (a non-covalent BTK inhibitor) with the covalent BTK inhibitors.

I would strongly encourage patients to participate in clinical trials so that we can better answer these important questions. When patients go online and read about MCL, they often see a median survival of 3 to 4 years, which is completely outdated. The overall prognosis of MCL has changed dramatically since I have been in the field. Hopefully, survival will continue to improve, and therapies will become more tolerable, as well.

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, 05/02/2023 - 12:00
Un-Gate On Date
Tue, 05/02/2023 - 12:00
Use ProPublica
CFC Schedule Remove Status
Tue, 05/02/2023 - 12:00
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
SF343187.5
Activity ID
95005
Product Name
Expert Interview Article Series
Product ID
106
Supporter Name /ID
Pirtobrutinib [ 5829 ]