Gene-expression profiling of bortezomib added to standard chemoimmunotherapy for diffuse large B-cell lymphoma (REMoDL-B): an open-label, randomised, phase 3 trial

Andrew Davies, Thomas E Cummin, Sharon Barrans, Tom Maishman, Christoph Mamot, Urban Novak, Josh Caddy, Louise Stanton, Shamim Kazmi-Stokes, Andrew McMillan, Paul Fields, Christopher Pocock, Graham P Collins, Richard Stephens, Francesco Cucco, Alexandra Clipson, Chulin Sha, Reuben Tooze, Matthew A Care, Gareth Griffiths, Ming-Qing Du, David R Westhead, Catherine Burton, Peter W M Johnson, Andrew Davies, Thomas E Cummin, Sharon Barrans, Tom Maishman, Christoph Mamot, Urban Novak, Josh Caddy, Louise Stanton, Shamim Kazmi-Stokes, Andrew McMillan, Paul Fields, Christopher Pocock, Graham P Collins, Richard Stephens, Francesco Cucco, Alexandra Clipson, Chulin Sha, Reuben Tooze, Matthew A Care, Gareth Griffiths, Ming-Qing Du, David R Westhead, Catherine Burton, Peter W M Johnson

Abstract

Background: Biologically distinct subtypes of diffuse large B-cell lymphoma can be identified using gene-expression analysis to determine their cell of origin, corresponding to germinal centre or activated B cell. We aimed to investigate whether adding bortezomib to standard therapy could improve outcomes in patients with these subtypes.

Methods: In a randomised evaluation of molecular guided therapy for diffuse large B-cell lymphoma with bortezomib (REMoDL-B), an open-label, adaptive, randomised controlled, phase 3 superiority trial, participants were recruited from 107 cancer centres in the UK (n=94) and Switzerland (n=13). Eligible patients had previously untreated, histologically confirmed diffuse large B-cell lymphoma with sufficient diagnostic material from initial biopsies for gene-expression profiling and pathology review; were aged 18 years or older; had ECOG performance status of 2 or less; had bulky stage I or stage II-IV disease requiring full-course chemotherapy; had measurable disease; and had cardiac, lung, renal, and liver function sufficient to tolerate chemotherapy. Patients initially received one 21-day cycle of standard rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone (R-CHOP; rituximab 375 mg/m2, cyclophosphamide 750 mg/m2, doxorubicin 50 mg/m2, and vincristine 1·4 mg/m2 [to a maximum of 2 mg total dose] intravenously on day 1 of the cycle, and prednisolone 100 mg orally once daily on days 1-5). During this time, we did gene-expression profiling using whole genome cDNA-mediated annealing, selection, extension, and ligation assay of tissue from routine diagnostic biopsy samples to determine the cell-of-origin subtype of each participant (germinal centre B cell, activated B cell, or unclassified). Patients were then centrally randomly assigned (1:1) via a web-based system, with block randomisation stratified by international prognostic index score and cell-of-origin subtype, to continue R-CHOP alone (R-CHOP group; control), or with bortezomib (RB-CHOP group; experimental; 1·3 mg/m2 intravenously or 1·6 mg/m2 subcutaneously) on days 1 and 8 for cycles two to six. If RNA extracted from the diagnostic tissues was of insufficient quality or quantity, participants were given R-CHOP as per the control group. The primary endpoint was 30-month progression-free survival, for the germinal centre and activated B-cell population. The primary analysis was on the modified intention-to-treat population of activated and germinal centre B-cell population. Safety was assessed in all participants who were given at least one dose of study drug. We report the progression-free survival and safety outcomes for patients in the follow-up phase after the required number of events occurred. This study was registered at ClinicalTrials.gov, number NCT01324596, and recruitment and treatment has completed for all participants, with long-term follow-up ongoing.

Findings: Between June 2, 2011, and June 10, 2015, 1128 eligible patients were registered, of whom 918 (81%) were randomly assigned to receive treatment (n=459 to R-CHOP, n=459 to RB-CHOP), comprising 244 (26·6%) with activated B-cell disease, 475 (51·7%) with germinal centre B cell disease, and 199 (21·7%) with unclassified disease. At a median follow-up of 29·7 months (95% CI 29·0-32·0), we saw no evidence for a difference in progression-free survival in the combined germinal centre and activated B-cell population between R-CHOP and RB-CHOP (30-month progression-free survival 70·1%, 95% CI 65·0-74·7 vs 74·3%, 69·3-78·7; hazard ratio 0·86, 95% CI 0·65-1·13; p=0·28). The most common grade 3 or worse adverse event was haematological toxicity, reported in 178 (39·8%) of 447 patients given R-CHOP and 187 (42·1%) of 444 given RB-CHOP. However, RB-CHOP was not associated with increased haematological toxicity and 398 [87·1%] of 459 participants assigned to receive RB-CHOP completed six cycles of treatment. Grade 3 or worse neuropathy occurred in 17 (3·8%) patients given RB-CHOP versus eight (1·8%) given R-CHOP. Serious adverse events occurred in 190 (42·5%) patients given R-CHOP, including five treatment-related deaths, and 223 (50·2%) given RB-CHOP, including four treatment-related deaths.

Interpretation: This is the first large-scale study in diffuse large B-cell lymphoma to use real-time molecular characterisation for prospective stratification, randomisation, and subsequent analysis of biologically distinct subgroups of patients. The addition of bortezomib did not improve progression-free survival.

Funding: Janssen-Cilag, Bloodwise, and Cancer Research UK.

Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Study profile ITT=intention-to-treat. R-CHOP=rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone. RB-CHOP=rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone.
Figure 2
Figure 2
Progression-free survival in the m ITT population (A), activated B-cell subgroup (B), germinal centre B-cell subgroup (C), and unclassified group (D), by treatment Data are for the mITT population, which comprises germinal centre and activated B-cell ITT participants (n=719); activated B-cell subgroup (n=244); germinal centre B-cell subgroup (n=475); and unclassified subgroup (n=199); with estimated proportions of participants achieving progression-free survival at 12 months and 30 months. HR=hazard ratio. ITT=intention-to-treat. mITT=modified ITT. R-CHOP=rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone. RB-CHOP=rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone.
Figure 3
Figure 3
Progression-free survival comparing double-hit lymphomas to non-rearranged cases (A) and double-expressor (high MYC and high BCL-2 mRNA) lymphomas to all other cases (B), by treatment group Data are progression-free survival and hazard ratio (HR), with non-DHL and non-DEL patients as reference categories. DEL=dual-expressor lymphoma. DHL=double-hit lymphoma. R-CHOP=rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone. RB-CHOP=rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone.
Figure 4
Figure 4
Forest plot of hazard ratios based on progression free survival for participants at high risk and with different molecular subtypes of disease, by treatment group Data are for all randomised participants (ie, ITT population). Hazard ratios and p values are effect estimates from a multivariable model adjusted for IPI score. DEL=dual-expressor lymphoma. DHL=double-hit lymphoma. IPI=international prognostic index. ITT=intention-to-treat. R-CHOP=rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone. RB-CHOP=rituximab, bortezomib, cyclophosphamide, doxorubicin, vincristine, and prednisolone.

References

    1. Coiffier B, Lepage E, Briere J. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med. 2002;346:235–242.
    1. Cunningham D, Hawkes EA, Jack A. Rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisolone in patients with newly diagnosed diffuse large B-cell non-Hodgkin lymphoma: a phase 3 comparison of dose intensification with 14-day versus 21-day cycles. Lancet. 2013;381:1817–1826.
    1. Smith A, Crouch S, Howell D, Burton C, Patmore R, Roman E. Impact of age and socioeconomic status on treatment and survival from aggressive lymphoma: a UK population-based study of diffuse large B-cell lymphoma. Cancer Epidemiol. 2015;39:1103–1112.
    1. van Imhoff GW, McMillan A, Matasar MJ. Ofatumumab versus rituximab salvage chemoimmunotherapy in relapsed or refractory diffuse large B-cell lymphoma: the ORCHARRD study. J Clin Oncol. 2016;35:544–551.
    1. Lenz G, Wright G, Dave SS. Stromal gene signatures in large-B-cell lymphomas. N Engl J Med. 2008;359:2313–2323.
    1. Young RM, Shaffer AL, 3rd, Phelan JD, Staudt LM. B-cell receptor signaling in diffuse large B-cell lymphoma. Semin Hematol. 2015;52:77–85.
    1. Dunleavy K, Pittaluga S, Czuczman MS. Differential efficacy of bortezomib plus chemotherapy within molecular subtypes of diffuse large B-cell lymphoma. Blood. 2009;113:6069–6076.
    1. Moreau P, Pylypenko H, Grosicki S. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study. Lancet Oncol. 2011;12:431–440.
    1. Cheson BD, Horning SJ, Coiffier B. Report of an international workshop to standardize response criteria for non-Hodgkin's lymphomas. J Clin Oncol. 1999;17:1244.
    1. Care MA, Barrans S, Worrillow L, Jack A, Westhead DR, Tooze RM. A microarray platform-independent classification tool for cell of origin class allows comparative analysis of gene expression in diffuse large B-cell lymphoma. PLoS One. 2013;8:e55895.
    1. Cucco F, Clipson A, Kennedy H. Mutation screening using formalin-fixed paraffin-embedded tissues: a stratified approach according to DNA quality. Lab Invest. 2018;98:1084–1092.
    1. Yuan J, Wright G, Rosenwald A. Identification of primary mediastinal large B-cell lymphoma at nonmediastinal sites by gene expression profiling. Am J Surg Pathol. 2015;39:1322–1330.
    1. Gómez H, Hidalgo M, Casanova L. Risk factors for treatment-related death in elderly patients with aggressive non-Hodgkin's lymphoma: results of a multivariate analysis. J Clin Oncol. 1998;16:2065–2069.
    1. Dubois S, Viailly PJ, Mareschal S. Next-generation sequencing in diffuse large B-cell lymphoma highlights molecular divergence and therapeutic opportunities: a LYSA study. Clin Cancer Res. 2016;22:2919–2928.
    1. Reddy A, Zhang J, Davis NS. Genetic and functional drivers of diffuse large B cell lymphoma. Cell. 2017;171:481–494.
    1. Johnson NA, Slack GW, Savage KJ. Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol. 2012;30:3452–3459.
    1. Scott DW, Wright GW, Williams PM. Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. Blood. 2014;123:1214–1217.
    1. Vitolo U, Trněný M, Belada D. Obinutuzumab or rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in previously untreated diffuse large B-cell lymphoma. J Clin Oncol. 2017;35:3529–3537.
    1. Leonard JP, Kolibaba KS, Reeves JA. Randomized phase II study of R-CHOP with or without bortezomib in previously untreated patients with non-germinal center B-cell-like diffuse large B-cell lymphoma. J Clin Oncol. 2017;35:3538–3546.
    1. Offner F, Samoilova O, Osmanov E. Frontline rituximab, cyclophosphamide, doxorubicin, and prednisone with bortezomib (VR-CAP) or vincristine (R-CHOP) for non-GCB DLBCL. Blood. 2015;126:1893–1901.
    1. Petrich AM, Gandhi M, Jovanovic B. Impact of induction regimen and stem cell transplantation on outcomes in double-hit lymphoma: a multicenter retrospective analysis. Blood. 2014;124:2354–2361.
    1. Clipson A, Barrans S, Zeng N. The prognosis of MYC translocation positive diffuse large B-cell lymphoma depends on the second hit. J Pathol Clin Res. 2015;1:125–133.
    1. Landsburg DJ, Falkiewicz MK, Maly J. Outcomes of patients with double-hit lymphoma who achieve first complete remission. J Clin Oncol. 2017;35:2260–2267.
    1. Maurer MJ, Ghesquieres H, Link BK. Diagnosis-to-treatment interval is an important clinical factor in newly diagnosed diffuse large B-cell lymphoma and has implication for bias in clinical trials. J Clin Oncol. 2018;36:1603–1610.
    1. Proctor IE, McNamara C, Rodriguez-Justo M, Isaacson PG, Ramsay A. Importance of expert central review in the diagnosis of lymphoid malignancies in a regional cancer network. J Clin Oncol. 2011;29:1431–1435.
    1. Stuhlmann-Laeisz C, Borchert A, Quintanilla-Martinez L. In Europe expression of EBNA2 is associated with poor survival in EBVpositive diffuse large B-cell lymphoma of the elderly. Leuk Lymphoma. 2016;57:39–44.
    1. Chapuy B, Stewart C, Dunford AJ. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24:679–690.
    1. Schmitz R, Wright GW, Huang DW. Genetics and pathogenesis of diffuse large B-cell lymphoma. N Engl J Med. 2018;378:1396–1407.

Source: PubMed

3
Se inscrever