Paul Gaynon

and 1 more

Clinical Relapse versus Treatment failureThe case for surveillance for reappearance of minimal measurable disease in pediatric patients with higher risk B-ALL.Paul S. Gaynon1 and Linwei Li21. University of Southern California, Keck School of Medicine, Los Angeles CA 900892. University of Texas, Rio Grande Valley School of Medicine, Edinburg, TX 78541Despite impressive progress in childhood acute lymphoblastic leukemia, an urgent clinical need remains. Patients still relapse and outcomes after relapse have changed little between 1996- 2006 and 2004 – 2014.1 Despite progress with late marrow relapse (1st remission (CR1) > 36 months), treatment of early relapse (CR1 < 36 months) remains unsatisfactory,2 especially in patients deemed higher risk at diagnosis, such as adolescents and young adults (AYA’s).3Progress to date derives from improved primary therapy. Further improvements in primary therapy have an ever-increasing price. Going from 50% to 70%, a 40% reduction in relapses, 5 patients need to be treated to prevent one relapse. Similarly, going from 75% to 85%, again a 40% reduction relapses, 10 patients need to be treated to prevent one relapse. With improving outcomes with primary therapy, we are facing an increasing number needed to treat for further improvement.4 All patients need to bear the burden of a novel intervention to benefit an ever smaller percentage. Not all interventions are successful.Recent experience suggests that we are reaching the limits of “intensification” of therapy, despite improvements in supportive care.5 For some patients such AYA’s, we may have surpassed tolerable limits. Querying the Pediatric Health Information system database, Gupta et al found a higher incidence of intensive care unit stays and increased toxicities in almost every organ system for AYA’s.6 Serious complications may prevent delivery of best care, resulting in relapse.Personalized molecularly targeted medicine is complicated by the vast interpatient diversity of ALL7 and intra-patient oligoclonality.8 Inhibition of a single driver pathway is unlikely sufficient for cure.9We have, however, newer modalities, such as inotuzumab,10,11 blinatumomab,12,13and chimeric antigen re-arranged (CAR) T-cells14,15that target large subsets of B-ALL. Blinatumomab has demonstrated value in relapsed B-ALL.12,13 Emerging experience has shown that blinatumomab and CAR T-cells work better at lower disease burdens.14-17 In 2017, blinatumomab was licensed for children and adults with B-ALL in 1st or second remission with measurable residual disease > 0.1%.18The presence of 25% marrow lymphoblasts blasts has long been the threshold for clinical marrow relapse. 19 Earlier reports showed that early detection of overt clinical relapse provides no clinical benefit.20,21 However, treatment fails, i.e., blast proliferation exceeds blast kill, prior to clinical relapse. We now have reliable technologies to identify low levels of re-appearing leukemia.22 Serial assessment of MRD and prompt intervention has yet to be tested systematically.In B-ALL, flow cytometry has largely overcome our inability to distinguish lymphoblasts from hematogones and recovery myeloblasts. Current flow cytometric or polymerase chain reaction technologies allows reliable detection of re-appearing lymphoblasts at levels 2 log10 below 1%.22 Next generation sequencing (NGS) allows detection 4 log10 below 1% and restaging of some patients called mistakenly classified MRD positive because of low numbers of low specificity targets.23 A new international consensus proposes that the confirmed presence of 1% lymphoblasts after the third month of therapy constitutes relapse or induction failure, if not preceded by a remission.24Flow cytometry, PCR, NGS, FISH, cytogenetics, and /or RT-PCR when relevant may be employed.Reappearance of MRD greater than some number predicts relapse in adult25 and pediatric trials.26Cheng et al report that reappearance of MRD reliably predicted relapse in their 30 patient cohort.27Early intervention may have clinical value. Wang et al reported on 1030 children who achieved MRD negative remission. One hundred fifty had MRD reappearance at a median time of 11 months. At 5 years, the EFS was 88.5% for continuously negative MRD and 38.4% for reemergent MRD. Eighty-five MRD reemergent patients subsequently relapsed at a median of 4.1 months. Reappearance of MRD was the most powerful adverse prognostic factor in multivariate analyses. An MRD cutoff of 0.15% gave the best discrimination. After reemergent MRD, 113 continued chemotherapy at their families’ choice and 37 underwent HSCT in CR1. The 2-year overall survival was 89% for HSCT and 46% for continued chemotherapy (p< 0.001); the cumulative incidence of relapse was 23% and 64% (p< 0.001).28MRD surveillance is not yet standard in pediatric ALL. However, MRD surveillance is already included in adult ALL guidelines.29,30 “Bone marrow aspirate can be considered as clinically indicated at a frequency of up to 3 to 6 months for at least 5 years.” 30In the past marrow sampling has been required as marrow and peripheral blood are unpredictably discordant in B-ALL. 31 Marrow aspiration is painful, and children often receive anesthesia. Marrow aspirates include a variable proportion of peripheral blood affecting the precision of MRD estimates. NGS may allow peripheral blood sampling.NGS has further lowered the limits of quantification to 10-6 and raised the possibility of peripheral blood monitoring in B-ALL.31 Rau et al found that end induction peripheral blood (PB) NGS was positive in nearly all cases marrow MRD by flow was positive at 0.01%. Muffly et al found that clinical relapse followed reappearance of peripheral blood NGS with a median of 90 days after HSCT and 60 days following CAR T therapy. Peripheral blood NGS surveillance of higher risk B-ALL patients seems worthy of investigation.Clinical features, molecular features, and response to therapy allow us to identify patients at greater or lesser risk of relapse.32 The adolescent and young adult population seems apt for a trial of such a strategy. Favorable cytogenetics are uncommon, and the marrow relapse rate is substantial.33 The burden of current therapy is already extreme and the efficacy of conventional salvage therapy poor.12Estimating a 20% relapse rate between 10 months and 36 months and q3month sampling, 8/10 patients would have 72 negative assays, and two relapsing patients might have 9 assays, for a total of 81 assays with about 1/40 being positive.Serial PB sampling adds little to the burden of treatment. Detection of confirmed treatment failure at an MRD level, allows immediate use of blinatumomab or CAR T-cells with a high probability to proceed to transplant MRD negative. Another round of toxic cytotoxic chemotherapy, much like the therapy that already failed, might be avoided. “If not now, when.”References1. Rheingold SR, Bhojwani D, Ji L, et al. Determinants of survival after first relapse of acute lymphoblastic leukemia: a Children’s Oncology Group study. Leukemia. 2024.2. Rheingold SR, Ji L, Xu X, et al. Prognostic factors for survival after relapsed acute lymphoblastic leukemia (ALL): A Children’s Oncology Group (COG) study. In: American Society of Clinical Oncology; 2019.3. Freyer DR, Devidas M, La M, et al. Postrelapse survival in childhood acute lymphoblastic leukemia is independent of initial treatment intensity: a report from the Children’s Oncology Group. Blood. 2011;117(11):3010-3015.4. Hasan H, Goddard K, Howard AF. Utility of the number needed to treat in paediatric haematological cancer randomised controlled treatment trials: a systematic review. BMJ open. 2019;9(2):e022839.5. van Binsbergen AL, de Haas V, van der Velden VHJ, de Groot-Kruseman HA, Fiocco MF, Pieters R. Efficacy and toxicity of high-risk therapy of the Dutch Childhood Oncology Group in childhood acute lymphoblastic leukemia. Pediatric blood & cancer. 2022;69(2):e29387.6. Gupta A, Damania RC, Talati R, O’Riordan MA, Matloub YH, Ahuja SP. Increased Toxicity Among Adolescents and Young Adults Compared with Children Hospitalized with Acute Lymphoblastic Leukemia at Children’s Hospitals in the United States. Journal of adolescent and young adult oncology. 2021;10(6):645-653.7. Chang TC, Chen W, Qu C, et al. Genomic Determinants of Outcome in Acute Lymphoblastic Leukemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2024:Jco2302238.8. Pieters R, Mullighan CG, Hunger SP. Advancing Diagnostics and Therapy to Reach Universal Cure in Childhood ALL. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2023;41(36):5579-5591.9. Kleppe M, Levine RL. Tumor heterogeneity confounds and illuminates: assessing the implications. Nature medicine. 2014;20(4):342-344.10. Dhillon S. Inotuzumab Ozogamicin: First Pediatric Approval.Paediatric drugs. 2024;26(4):459-467.11. O’Brien MM, Ji L, Shah NN, et al. Phase II Trial of Inotuzumab Ozogamicin in Children and Adolescents With Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia: Children’s Oncology Group Protocol AALL1621. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2022;40(9):956-967.12. Brown PA, Ji L, Xu X, et al. Effect of Postreinduction Therapy Consolidation With Blinatumomab vs Chemotherapy on Disease-Free Survival in Children, Adolescents, and Young Adults With First Relapse of B-Cell Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. Jama. 2021;325(9):833-842.13. Hogan LE, Brown PA, Ji L, et al. Children’s Oncology Group AALL1331: Phase III Trial of Blinatumomab in Children, Adolescents, and Young Adults With Low-Risk B-Cell ALL in First Relapse. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2023;41(25):4118-4129.14. Lamble AJ, Myers RM, Taraseviciute A, et al. Preinfusion factors impacting relapse immunophenotype following CD19 CAR T cells.Blood advances. 2023;7(4):575-585.15. Park JH, Rivière I, Gonen M, et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. The New England journal of medicine. 2018;378(5):449-459.16. Gökbuget N, Dombret H, Bonifacio M, et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood. 2018;131(14):1522-1531.17. Queudeville M, Stein AS, Locatelli F, et al. Low leukemia burden improves blinatumomab efficacy in patients with relapsed/refractory B-cell acute lymphoblastic leukemia. Cancer. 2023;129(9):1384-1393.18. Jen EY, Xu Q, Schetter A, et al. FDA Approval: Blinatumomab for Patients with B-cell Precursor Acute Lymphoblastic Leukemia in Morphologic Remission with Minimal Residual Disease. Clinical cancer research : an official journal of the American Association for Cancer Research. 2019;25(2):473-477.19. Mastrangelo R, Poplack D, Bleyer A, Riccardi R, Sather H, D’Angio G. Report and recommendations of the Rome workshop concerning poor-prognosis acute lymphoblastic leukemia in children: biologic bases for staging, stratification, and treatment. Medical and pediatric oncology. 1986;14(3):191-194.20. Rogers PC, Bleyer WA, Coccia P, et al. Yield of unpredicted bone-marrow relapse diagnosed by routine marrow aspiration in children with acute lymphoblastic leukaemia. A report from the Children’s Cancer Study Group. Lancet (London, England). 1984;1(8390):1320-1322.21. Rubnitz JE, Hijiya N, Zhou Y, Hancock ML, Rivera GK, Pui CH. Lack of benefit of early detection of relapse after completion of therapy for acute lymphoblastic leukemia. Pediatric blood & cancer. 2005;44(2):138-141.22. Contreras Yametti GP, Ostrow TH, Jasinski S, Raetz EA, Carroll WL, Evensen NA. Minimal Residual Disease in Acute Lymphoblastic Leukemia: Current Practice and Future Directions. Cancers. 2021;13(8).23. Svaton M, Skotnicova A, Reznickova L, et al. NGS better discriminates true MRD positivity for the risk stratification of childhood ALL treated on an MRD-based protocol. Blood. 2023;141(5):529-533.24. Buchmann S, Schrappe M, Baruchel A, et al. Remission, treatment failure, and relapse in pediatric ALL: an international consensus of the Ponte-di-Legno Consortium. Blood. 2022;139(12):1785-1793.25. Raff T, Gökbuget N, Lüschen S, et al. Molecular relapse in adult standard-risk ALL patients detected by prospective MRD monitoring during and after maintenance treatment: data from the GMALL 06/99 and 07/03 trials. Blood. 2007;109(3):910-915.26. Paganin M, Fabbri G, Conter V, et al. Postinduction minimal residual disease monitoring by polymerase chain reaction in children with acute lymphoblastic leukemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014;32(31):3553-3558.27. Cheng S, Inghirami G, Cheng S, Tam W. Simple deep sequencing-based post-remission MRD surveillance predicts clinical relapse in B-ALL.Journal of hematology & oncology. 2018;11(1):105.28. Wang Y, Xue YJ, Jia YP, Zuo YX, Lu AD, Zhang LP. Re-Emergence of Minimal Residual Disease Detected by Flow Cytometry Predicts an Adverse Outcome in Pediatric Acute Lymphoblastic Leukemia. Frontiers in oncology. 2020;10:596677.29. Gökbuget N, Boissel N, Chiaretti S, et al. Management of ALL in adults: 2024 ELN recommendations from a European expert panel.Blood. 2024;143(19):1903-1930.30. Network NCC. Acute Lymphoblastic Leukemia (Version 2.2024) 2024.31. Pierce E, Mautner B, Mort J, et al. MRD in ALL: Optimization and Innovations. Current hematologic malignancy reports. 2022;17(4):69-81.32. Hunger SP, Loh ML, Whitlock JA, et al. Children’s Oncology Group’s 2013 blueprint for research: acute lymphoblastic leukemia.Pediatric blood & cancer. 2013;60(6):957-963.33. Burke MJ, Devidas M, Chen Z, et al. Outcomes in adolescent and young adult patients (16 to 30 years) compared to younger patients treated for high-risk B-lymphoblastic leukemia: report from Children’s Oncology Group Study AALL0232. Leukemia. 2022;36(3):648-655.

Sebastian de Armas

and 7 more

Background: ABVD, the standard-of-care in adult Hodgkin lymphoma (HL), has not been directly compared to ABVE-PC, a pediatric regimen designed to reduce late-effects. We aimed to compare the effectiveness and associated toxicities of these two regimens used in the same institution. Methods: This retrospective cohort study evaluated a total of 224 patients diagnosed with HL between 1999 and 2018 at Children’s Hospital Los Angeles (CHLA), of which 93 patients were eligible having received ABVD (n=46) or ABVE-PC (n=47) chemotherapy as their initial treatment. Descriptive analyses were performed using the Student’s t-test or Fisher’s exact test. Survival analysis used the Kaplan-Meier method. Events included: death, relapse, secondary malignancy, need for radiation therapy, pulmonary toxicity and cardiomyopathy determined by shortening fraction <29%. Analyses followed an intention-to-treat principle. Results: There was no difference in baseline characteristics between the patients receiving ABVE-PC or ABVD in regard for stage, risk group or prognostic variables, such as the presence or absence of “B” symptoms, bulky disease, and extra-nodal involvement. A greater proportion of patients treated with ABVE-PC received consolidating external beam radiation treatment (XRT) either by randomization or by response compared to ABVD (59.6% vs 32.6% respectively, p=0.01). While not statistically significant, response to therapy, assessed by PET/CT where available, mirrored the need for radiation (rapid response 58.3% vs 90.0%, n=34, p=0.11). There was no difference in event-free survival (p=0.63) or overall survival (p=0.37) with a median follow up length of 3.9 years. Conclusion: ABVD and ABVE-PC achieved similar survival outcomes in our single-institution cohort