Role of stem cell transplant in CD30+ PTCL following frontline brentuximab vedotin plus CHP or CHOP in ECHELON-2

Kerry J Savage, Steven M Horwitz, Ranjana Advani, Jacob Haaber Christensen, Eva Domingo-Domenech, Giuseppe Rossi, Franck Morschhauser, Onder Alpdogan, Cheolwon Suh, Kensei Tobinai, Andrei Shustov, Marek Trneny, Sam Yuen, Pier Luigi Zinzani, Lorenz Trümper, Tim Ilidge, Owen A O'Connor, Barbara Pro, Harry Miao, Veronica Bunn, Keenan Fenton, Michelle Fanale, Markus Puhlmann, Swaminathan Iyer, Kerry J Savage, Steven M Horwitz, Ranjana Advani, Jacob Haaber Christensen, Eva Domingo-Domenech, Giuseppe Rossi, Franck Morschhauser, Onder Alpdogan, Cheolwon Suh, Kensei Tobinai, Andrei Shustov, Marek Trneny, Sam Yuen, Pier Luigi Zinzani, Lorenz Trümper, Tim Ilidge, Owen A O'Connor, Barbara Pro, Harry Miao, Veronica Bunn, Keenan Fenton, Michelle Fanale, Markus Puhlmann, Swaminathan Iyer

Abstract

Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of aggressive non-Hodgkin lymphomas, the majority of which have high relapse rates following standard therapy. Despite use of consolidative stem cell transplant (SCT) following frontline therapy, there remains no consensus on its utility. The double-blind randomized phase 3 ECHELON-2 study (#NCT01777152; clinicaltrials.gov) demonstrated improved progression-free survival (PFS) and overall survival with frontline brentuximab vedotin plus cyclophosphamide, doxorubicin, and prednisone (A+CHP). Herein, we conducted an exploratory subgroups analysis of the impact of consolidative SCT on PFS in patients with previously untreated CD30+ PTCL (ALK- anaplastic large cell lymphoma [ALCL] and non-ALCL) who were in complete response (CR) after frontline treatment with A+CHP or cyclophosphamide, doxorubicin, vincristine, and prednisone. Median PFS follow-up was 47.57 months. The PFS hazard ratio was 0.36, equating to a 64% reduction in the risk of a PFS event in patients who underwent SCT. The median PFS in patients who underwent SCT was not reached, vs 55.66 months in patients who did not undergo SCT. PFS results favored the use of SCT in both ALK- ALCL and non-ALCL subgroups. These data support the consideration of consolidative SCT in patients with CD30+PTCL who achieve CR following treatment with A+CHP.

Conflict of interest statement

Conflict-of-interest disclosure: S.M.H. has consulted, received honorarium from, or participated in advisory boards for Acrotech Biopharma, C4 Therapeutics, Janssen, Kura Oncology, Kyowa Hakko Kirin, Myeloid Therapeutics, ONO Pharmaceuticals, Seagen Inc., Takeda, Trillium Therapeutics, and Vividion Therapeutics and has received research support for clinical trials from ADC Therapeutics, Affimed, Aileron, Celgene, Daiichi Sankyo, Forty Seven Inc., Kyowa Hakko Kirin, Millennium/Takeda, Portola Pharmaceuticals, Seagen Inc., Trillium Therapeutics, and Verastem. T.I. serves on the speakers bureau for Takeda. S.I. receives research funding from Arog, Bristol-Myers Squibb, Genentech/Roche, Incyte, and Seagen Inc. R.A. consults for Astra Zeneca, Bayer Healthcare Pharmaceuticals, Cell Medica, Celgene, Genentech/Roche, Gilead, KitePharma, Kyowa, Portola Pharmaceuticals, Sanofi, Seagen Inc., and Takeda. J.H.A. receives research funding from Seagen Inc. F.M. serves on the board of directors or advisory committees at Abbivie, Celgene, Epizyme, Gilead; receives honoraria from Janssen; consultants with and receives honoraria from and serves on the board of directors at Roche; and consultants for Servier. E.D.-D. consults for and receives travel, accommodation, expense, and research funding from Takeda; received travel and research funding from Bristol-Myers Squibb; receives travel, accommodation, and expense funding from Roche and Janssen; and receives research funding from Seagen Inc. G.R. consults for and receives honoraria from Daiichi Sankyo; serves on the advisory board for Roche, Janssen, Celgene, Amgen, Novartis, and Astellas; serves on the advisory board and receives travel expense funding from Gilead and Alexion; receives honoraria from and serves on the advisory board for Sandoz; receives honoraria from Incyte, Sanofi, Bristol-Myers Squibb, Mundipharma, and Novartis; is a member of the board of directors or advisory committees at Abbvie, Jazz, Pfizer, and Teva; receives research funding from Seagen Inc.; and travel expenses from Alexion. K.T. receives honoraria from Bristol-Myers Squibb, Eisai, and Solasia; consults and receives honoraria from Celgene, Chugai Pharma, Daiichi Sankyo, HUYA Bioscience, Kyowa Kirin, Mundipharma, Ono Pharma, Takeda, Yakult, and Zenyaku Kogyo; and consults for SymBio. A.R.S. receives research funding from Seagen Inc. K.J.S. consults and receives honoraria from Seagen Inc., Bristol Myers Squibb, Merck, Janssen, Kyowa; is a member of the advisory committees at Bristol-Myers Squibb, Steering Committee Beigene, and DSMC Regeneron; and receives institutional research funding from Roche and BMS. P.L.Z. receives honoraria and is a member of the board of directors or advisory committees and speakers bureau at Merck, AbbVie, Takeda, Incyte, Kirin Kyowa, and ADC Therapeutics; consults for, receives honoraria from, and is a member of the board of directors or advisory committees and speakers bureau at BMS, Gilead, Roche, Servier, EUSA Pharma, Verastem, Celltrion, Janssen-Cilag, Sandoz, MSD, Immune Design, and Celgene; receives honoraria from and is on the speakers bureau at TG Therapeutics Inc.; consults for and is a member of the board of directors or advisory committees at Sanofi; consults for and is a member of the board of directors or advisory committees, receives research funding, and is on the speakers bureau at Portola; consults for and is on the speakers bureau at EUSA Pharma and Kyowa Kirin; and is a member of the board of directors or advisory committees and speakers bureau at Immune Design. H.M. is a current equity holder at Takeda. V.B. receives research funding from Seagen Inc. and is currently employed at Takeda. K.F. is an employee of an has an equity interest in Seagen Inc. M.A.F. is an employee of and has an equity interest in Seagen Inc. M.P. is an employee of and has an equity interest in Seagen Inc. L.T. consults for Janssen and Nordic Nanovector; receives research funding from Mundipharma, Roche, and Seagen Inc.; and consults for and receives research funding from Takeda Europe. O.A. serves on the advisory board at Kyowa Kirin and Seagen Inc. and receives research funding from Seagen Inc. C.S. receives research funding from Seagen Inc. M.T. receives research funding from Seagen Inc. The remaining authors declare no competing financial interests.

The current affiliation for A.S. is Seagen Inc., Bothell, WA.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Figure 1.
Figure 1.
Patient flow diagram for ECHELON-2 exploratory analysis: A+CHP arm. There were no significant differences (P = .49) in the baseline international prognostic index score distribution between patients who received SCT and patients who did not receive SCT. Note that 12 transplanted patients were excluded from the analysis (4 patients with ALK+; 8 did not have CR at EOT [6 PR, 1 SD, and 1 not evaluable]), for a total analysis population of n = 38. Consistent criteria were applied per protocol for response assessments. PR, partial response.
Figure 2.
Figure 2.
PFS by consolidative SCT: A+CHP arm. (A) PFS by consolidative SCT after A+CHP in patients with CR at EOT: ALK− sALCL and Non-sALCL. (B) PFS by consolidative SCT after A+CHP in patients with CR at EOT: ALK− sALCL. (C) PFS by consolidative SCT after A+CHP in patients with CR at EOT: Non-sALCL. ITT, intention to transplant.

References

    1. Anderson JR, Armitage JO, Weisenburger DD. Epidemiology of the non-Hodgkin’s lymphomas: distributions of the major subtypes differ by geographic locations. Non-Hodgkin’s Lymphoma Classification Project. Ann Oncol. 1998;9(7):717–720.
    1. d’Amore F, Gaulard P, Trümper L, et al. ESMO Guidelines Committee Peripheral T-cell lymphomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(suppl 5):v108–v115.
    1. Horwitz SM, Zelenetz AD, Gordon LI, et al. NCCN Guidelines insights: non-Hodgkin’s lymphomas, version 3.2016. J Natl Compr Canc Netw. 2016;14(9):1067–1079.
    1. Vose J, Armitage J, Weisenburger D, International T-Cell Lymphoma Project International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes. J Clin Oncol. 2008;26(25):4124–4130.
    1. Savage KJ, Chhanabhai M, Gascoyne RD, Connors JM. Characterization of peripheral T-cell lymphomas in a single North American institution by the WHO classification. Ann Oncol. 2004;15(10):1467–1475.
    1. Simon A, Peoch M, Casassus P, et al. Upfront VIP-reinforced-ABVD (VIP-rABVD) is not superior to CHOP/21 in newly diagnosed peripheral T cell lymphoma. Results of the randomized phase III trial GOELAMS-LTP95. Br J Haematol. 2010;151(2):159–166.
    1. Savage KJ, Harris NL, Vose JM, et al. International Peripheral T-Cell Lymphoma Project ALK- anaplastic large-cell lymphoma is clinically and immunophenotypically different from both ALK+ ALCL and peripheral T-cell lymphoma, not otherwise specified: report from the International Peripheral T-Cell Lymphoma Project. Blood. 2008;111(12):5496–5504.
    1. Bossard C, Dobay MP, Parrens M, et al. Immunohistochemistry as a valuable tool to assess CD30 expression in peripheral T-cell lymphomas: high correlation with mRNA levels. Blood. 2014;124(19):2983–2986.
    1. Sabattini E, Pizzi M, Tabanelli V, et al. CD30 expression in peripheral T-cell lymphomas. Haematologica. 2013;98(8):e81–e82.
    1. Savage KJ. Prognosis and primary therapy in peripheral T-cell lymphomas. Hematology Am Soc Hematol Educ Program. 2008;20081:280–288.
    1. d’Amore F, Relander T, Lauritzsen GF, et al. Up-front autologous stem-cell transplantation in peripheral T-cell lymphoma: NLG-T-01. J Clin Oncol. 2012;30(25):3093–3099.
    1. Smith SM, Burns LJ, van Besien K, et al. Hematopoietic cell transplantation for systemic mature T-cell non-Hodgkin lymphoma. J Clin Oncol. 2013;31(25):3100–3109.
    1. Abramson JS, Feldman T, Kroll-Desrosiers AR, et al. Peripheral T-cell lymphomas in a large US multicenter cohort: prognostication in the modern era including impact of frontline therapy. Ann Oncol. 2014;25(11):2211–2217.
    1. Fossard G, Broussais F, Coelho I, et al. Role of up-front autologous stem-cell transplantation in peripheral T-cell lymphoma for patients in response after induction: an analysis of patients from LYSA centers. Ann Oncol. 2018;29(3):715–723. [published correction appears in Ann Oncol. 2021;32(7):945]
    1. Horwitz S, O’Connor OA, Pro B, et al. ECHELON-2 Study Group Brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2): a global, double-blind, randomised, phase 3 trial. Lancet. 2019;393(10168):229–240.
    1. Horwitz S, O'Connor OA, Pro B, et al. The ECHELON-2 trial: 5-year results of a randomized, phase 3 study of brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma. Ann Oncol. 2022;33(3):288–298.
    1. Cheson BD, Pfistner B, Juweid ME, et al. International Harmonization Project on Lymphoma Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579–586.
    1. Pedersen MB, Hamilton-Dutoit SJ, Bendix K, et al. DUSP22 and TP63 rearrangements predict outcome of ALK-negative anaplastic large cell lymphoma: a Danish cohort study. Blood. 2017;130(4):554–557.
    1. Hapgood G, Ben-Neriah S, Mottok A, et al. Identification of high-risk DUSP22-rearranged ALK-negative anaplastic large cell lymphoma. Br J Haematol. 2019;186(3):e28–e31.

Source: PubMed

3
Tilaa