Circulating tumor DNA predicts therapeutic outcome in mantle cell lymphoma

Rahul Lakhotia, Christopher Melani, Kieron Dunleavy, Stefania Pittaluga, Nakhle Saba, Liza Lindenberg, Esther Mena, Ethan Bergvall, Andrea Nicole Lucas, Allison Jacob, Erik Yusko, Seth M Steinberg, Elaine S Jaffe, Adrian Wiestner, Wyndham H Wilson, Mark Roschewski, Rahul Lakhotia, Christopher Melani, Kieron Dunleavy, Stefania Pittaluga, Nakhle Saba, Liza Lindenberg, Esther Mena, Ethan Bergvall, Andrea Nicole Lucas, Allison Jacob, Erik Yusko, Seth M Steinberg, Elaine S Jaffe, Adrian Wiestner, Wyndham H Wilson, Mark Roschewski

Abstract

Mantle cell lymphoma (MCL) is biologically and clinically heterogeneous and would benefit from prognostic biomarkers to guide management. Circulating tumor DNA (ctDNA) is a novel prognostic biomarker in diffuse large B-cell lymphoma that may have applicability in MCL. We analyzed ctDNA dynamics in previously untreated patients with MCL who received induction therapy with bortezomib and DA-EPOCH-R for 6 cycles followed by random assignment to observation or bortezomib maintenance in responding patients in a prospective phase 2 study. Most patients also underwent initial treatment window of bortezomib alone prior to induction. Serum was collected pretreatment, after the window, after cycles 1 and 2, at the end of induction, and at each follow-up visit along with restaging computed tomography scans. Next-generation sequencing was used to identify and quantify ctDNA encoding the immunoglobulin receptor sequences in serum as markers of minimal residual disease. Fifty-three patients were enrolled, with a median follow-up of 12.7 years. Patients without detectable ctDNA after 2 cycles of induction had longer progression-free survival (PFS) and overall survival (OS) compared with those with detectable ctDNA (median PFS, 2.7 vs 1.8 years; overall P = .005; median OS, 13.8 vs 7.4 years; overall P = .03). Notably, in vivo assessment of ctDNA dynamics during the bortezomib window was not prognostic, and there was no difference in PFS or OS with bortezomib maintenance. ctDNA monitoring after induction showed that molecular relapse preceded clinical relapse in some cases. In conclusion, interim ctDNA negativity strongly correlates with improved survival and supports the investigation of response-adapted strategies. This trial was registered at www.clinicaltrials.gov as #NCT00114738.

© 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

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
CONSORT diagram. Patients first received bortezomib as monotherapy in a window for 21 days, except for those with rapidly progressive disease, who proceeded directly to induction therapy. Patients who responded to induction and did not have significant peripheral neuropathy were randomly assigned to bortezomib maintenance or initial observation. DA-EPOCH-R, dose-adjusted etoposide, doxorubicin, and cyclophosphamide with prednisone, vincristine, and rituximab; CNS, central nervous system.
Figure 2.
Figure 2.
Kaplan-Meier estimates of PFS and OS of patients enrolled with untreated MCL. Median follow-up was 12.7 years. (A) The 5-year PFS of all patients was 23.1% (95% CI, 13-35). (B) The 5-year OS of all patients was 78.3% (95% CI, 64-87). (C) The 5-year PFS of patients randomly assigned to bortezomib maintenance vs observation was 14.3% (95% CI, 2-37) vs 37.5% (95% CI, 15-60; P = .39). (D) The 5-year OS of patients randomly assigned to bortezomib maintenance vs observation was 69.2% (95% CI, 37-87) vs 87.5% (95% CI, 59-97; P = .32).
Figure 3.
Figure 3.
Pretreatment ctDNA is associated with other prognostic variables. (A) Scatter plot shows the median pretreatment ctDNA for all patients and the relationship of pretreatment ctDNA in classic histology compared with blastoid histology. (B) Scatter plot shows pretreatment ctDNA across MIPI categories. (C) Scatter plot demonstrates the relationship of pretreatment ctDNA and the Ki67 proliferation index. (D) Spearman correlation of pretreatment ctDNA and SUVmax on baseline FDG-PET scan. (E) Spearman correlation of pretreatment ctDNA and TMTV on baseline FDG-PET scan. (F) Kaplan-Meier estimates of PFS in patients above the median (ctDNA high) compared with below the median (ctDNA low). *P < .05.
Figure 4.
Figure 4.
Circulating tumor DNA dynamics during induction therapy. (A) Qualitative ctDNA response to induction therapy after 1 cycle, after 2 cycles, and at the end of induction therapy. (B) Quantitative ctDNA response after the bortezomib window and after 1 cycle, after 2 cycles, and at the end of induction therapy.
Figure 5.
Figure 5.
Kaplan-Meier estimates of PFS and OS based on ctDNA detection at landmark time points during induction therapy. (A) PFS in patients with no detectable ctDNA (blue) compared with patients with detectable ctDNA (red) at the end of induction therapy. (B) OS in patients with no detectable ctDNA (blue) compared with patients with detectable ctDNA (red) at the end of induction therapy. (C) PFS in patients with no detectable ctDNA (blue) compared with patients with detectable ctDNA (red) after 2 cycles of induction therapy. (D) OS in patients with no detectable ctDNA (blue) compared with patients with detectable ctDNA (red) after 2 cycles of induction therapy. (E) PFS in patients with no detectable ctDNA (blue) compared with patients with detectable ctDNA (red) after 1 cycle of induction therapy. (F) OS in patients with no detectable ctDNA (blue) compared with patients with detectable ctDNA (red) after 1 cycle of induction therapy.
Figure 6.
Figure 6.
Circulating tumor DNA after induction therapy. Patients (n = 47) who responded to induction therapy and proceeded to part C of therapy (maintenance or observation) were included. Serum collected at protocol-defined time points was analyzed blinded to clinical outcomes for the detection of ctDNA (red circle) or absence of detectable ctDNA (green circle). Six patients (blue shade and above bold line) had no detectable ctDNA at end of induction therapy (EOT) and were without radiologic progression at the time of data cutoff. Thirty-four patients (below bold line and above dotted line) had no detectable ctDNA at EOT and were monitored until radiologic progression (square). Seven patients (below dotted line) had detectable ctDNA at EOT and were monitored until radiologic progression. Twenty-eight patients (red shade) progressed and had ctDNA positivity at the time of or before progression, whereas 13 patients (green shade) progressed with no prior detectable ctDNA. 1C, end of the first cycle of induction therapy; 2C, end of the second cycle of induction therapy.

References

    1. Pérez-Galán P, Dreyling M, Wiestner A. Mantle cell lymphoma: biology, pathogenesis, and the molecular basis of treatment in the genomic era. Blood. 2011;117(1):26-38.
    1. Martin P, Chadburn A, Christos P, et al. . Outcome of deferred initial therapy in mantle-cell lymphoma. J Clin Oncol. 2009;27(8):1209-1213.
    1. Romaguera JE, Fayad L, Rodriguez MA, et al. . High rate of durable remissions after treatment of newly diagnosed aggressive mantle-cell lymphoma with rituximab plus hyper-CVAD alternating with rituximab plus high-dose methotrexate and cytarabine. J Clin Oncol. 2005;23(28):7013-7023.
    1. Hermine O, Hoster E, Walewski J, et al. ; European Mantle Cell Lymphoma Network . Addition of high-dose cytarabine to immunochemotherapy before autologous stem-cell transplantation in patients aged 65 years or younger with mantle cell lymphoma (MCL Younger): a randomised, open-label, phase 3 trial of the European Mantle Cell Lymphoma Network. Lancet. 2016;388(10044):565-575.
    1. Geisler CH, Kolstad A, Laurell A, et al. ; Nordic Lymphoma Group . Long-term progression-free survival of mantle cell lymphoma after intensive front-line immunochemotherapy with in vivo-purged stem cell rescue: a nonrandomized phase 2 multicenter study by the Nordic Lymphoma Group. Blood. 2008;112(7):2687-2693.
    1. Eve HE, Furtado MV, Hamon MD, Rule SA. Time to treatment does not influence overall survival in newly diagnosed mantle-cell lymphoma. J Clin Oncol. 2009;27(32):e189-e190, author reply e191.
    1. Kluin-Nelemans HC, Hoster E, Hermine O, et al. . Treatment of older patients with mantle-cell lymphoma. N Engl J Med. 2012;367(6):520-531.
    1. Le Gouill S, Thieblemont C, Oberic L, et al. ; LYSA Group . Rituximab after autologous stem-cell transplantation in mantle-cell lymphoma. N Engl J Med. 2017;377(13):1250-1260.
    1. Eskelund CW, Kolstad A, Jerkeman M, et al. . 15-year follow-up of the Second Nordic Mantle Cell Lymphoma trial (MCL2): prolonged remissions without survival plateau. Br J Haematol. 2016;175(3):410-418.
    1. Rosenwald A, Wright G, Wiestner A, et al. . The proliferation gene expression signature is a quantitative integrator of oncogenic events that predicts survival in mantle cell lymphoma. Cancer Cell. 2003;3(2):185-197.
    1. Scott DW, Abrisqueta P, Wright GW, et al. ; Lymphoma/Leukemia Molecular Profiling Project . New molecular assay for the proliferation signature in mantle cell lymphoma applicable to formalin-fixed paraffin-embedded biopsies. J Clin Oncol. 2017;35(15):1668-1677.
    1. Clot G, Jares P, Giné E, et al. . A gene signature that distinguishes conventional and leukemic nonnodal mantle cell lymphoma helps predict outcome. Blood. 2018;132(4):413-422.
    1. Nadeu F, Martin-Garcia D, Clot G, et al. . Genomic and epigenomic insights into the origin, pathogenesis, and clinical behavior of mantle cell lymphoma subtypes. Blood. 2020;136(12):1419-1432.
    1. Palacios A, Lopez A, Salar A, Cervera M, Gironella M. Rituximab-EPOCH, an effective front-line therapy for mantle cell lymphoma [abstract]. Blood. 2007;110(11):XXX. Abstract 4490.
    1. Neelapu SS, Kwak LW, Kobrin CB, et al. . Vaccine-induced tumor-specific immunity despite severe B-cell depletion in mantle cell lymphoma. Nat Med. 2005;11(9):986-991.
    1. Robak T, Huang H, Jin J, et al. ; LYM-3002 Investigators . Bortezomib-based therapy for newly diagnosed mantle-cell lymphoma. N Engl J Med. 2015;372(10):944-953.
    1. Robak T, Jin J, Pylypenko H, et al. ; LYM-3002 investigators . Frontline bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone (VR-CAP) versus rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) in transplantation-ineligible patients with newly diagnosed mantle cell lymphoma: final overall survival results of a randomised, open-label, phase 3 study. Lancet Oncol. 2018;19(11):1449-1458.
    1. Till BG, Li H, Bernstein SH, et al. . Phase II trial of R-CHOP plus bortezomib induction therapy followed by bortezomib maintenance for newly diagnosed mantle cell lymphoma: SWOG S0601. Br J Haematol. 2016;172(2):208-218.
    1. Drach J, Huang H, Samoilova O, et al. . Efficacy and safety of frontline rituximab, cyclophosphamide, doxorubicin and prednisone plus bortezomib (VR-CAP) or vincristine (R-CHOP) in a subset of newly diagnosed mantle cell lymphoma patients medically eligible for transplantation in the randomized, phase 3 LYM-3002 study. Leuk Lymphoma. 2018;59(4):896-903.
    1. Gressin R, Daguindau N, Tempescul A, et al. ; Lymphoma Study Association . A phase 2 study of rituximab, bendamustine, bortezomib and dexamethasone for first-line treatment of older patients with mantle cell lymphoma. Haematologica. 2019;104(1):138-146.
    1. Verhoef G, Robak T, Huang H, et al. . Association between quality of response and outcomes in patients with newly diagnosed mantle cell lymphoma receiving VR-CAP versus R-CHOP in the phase 3 LYM-3002 study. Haematologica. 2017;102(5):895-902.
    1. Kaplan LD, Maurer MJ, Stock W, et al. . Bortezomib consolidation or maintenance following immunochemotherapy and autologous stem cell transplantation for mantle cell lymphoma: CALGB/Alliance 50403. Am J Hematol. 2020;95(6):583-593.
    1. Doorduijn JK, Zijlstra JM, Lugtenburg PJ, et al. . Bortezomib maintenance after R-CHOP, cytarabine and autologous stem cell transplantation in newly diagnosed patients with mantle cell lymphoma, results of a randomised phase II HOVON trial. Br J Haematol. 2020;190(3):385-393.
    1. Pott C, Schrader C, Gesk S, et al. . Quantitative assessment of molecular remission after high-dose therapy with autologous stem cell transplantation predicts long-term remission in mantle cell lymphoma. Blood. 2006;107(6):2271-2278.
    1. Pott C, Hoster E, Delfau-Larue MH, et al. . Molecular remission is an independent predictor of clinical outcome in patients with mantle cell lymphoma after combined immunochemotherapy: a European MCL intergroup study. Blood. 2010;115(16):3215-3223.
    1. Liu H, Johnson JL, Koval G, et al. . Detection of minimal residual disease following induction immunochemotherapy predicts progression free survival in mantle cell lymphoma: final results of CALGB 59909. Haematologica. 2012;97(4):579-585.
    1. Kolstad A, Pedersen LB, Eskelund CW, et al. ; Nordic Lymphoma Group . Molecular monitoring after autologous stem cell transplantation and preemptive rituximab treatment of molecular relapse; results from the Nordic Mantle Cell Lymphoma Studies (MCL2 and MCL3) with median follow-up of 8.5 years. Biol Blood Marrow Transplant. 2017;23(3):428-435.
    1. Guidot DM, Switchenko JM, Nastoupil LJ, et al. . Surveillance imaging in mantle cell lymphoma in first remission lacks clinical utility. Leuk Lymphoma. 2018;59(4):888-895.
    1. Roschewski M, Dunleavy K, Pittaluga S, et al. . Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16(5):541-549.
    1. Kurtz DM, Green MR, Bratman SV, et al. . Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679-3687.
    1. Armand P, Redd R, Bsat J, et al. . A phase 2 study of Rituximab-Bendamustine and Rituximab-Cytarabine for transplant-eligible patients with mantle cell lymphoma. Br J Haematol. 2016;173(1):89-95.
    1. Ladetto M, Brüggemann M, Monitillo L, et al. . Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia. 2014;28(6):1299-1307.
    1. Kurtz DM, Scherer F, Jin MC, et al. . Circulating tumor DNA measurements as early outcome predictors in diffuse large B-cell lymphoma. J Clin Oncol. 2018;36(28):2845-2853.
    1. Agarwal R, Chan YC, Tam CS, et al. . Dynamic molecular monitoring reveals that SWI-SNF mutations mediate resistance to ibrutinib plus venetoclax in mantle cell lymphoma. Nat Med. 2019;25(1):119-129.
    1. Kurtz DM, Esfahani MS, Scherer F, et al. . Dynamic risk profiling using serial tumor biomarkers for personalized outcome prediction. Cell. 2019;178(3):699-713.e19.
    1. Wilson WH, Dunleavy K, Pittaluga S, et al. . Phase II study of dose-adjusted EPOCH and rituximab in untreated diffuse large B-cell lymphoma with analysis of germinal center and post-germinal center biomarkers. J Clin Oncol. 2008;26(16):2717-2724.
    1. Cheson BD, Horning SJ, Coiffier B, et al. ; NCI Sponsored International Working Group . Report of an international workshop to standardize response criteria for non-Hodgkin’s lymphomas [published correction appears in J Clin Oncol. 2000;18(11):2351]. J Clin Oncol. 1999;17(4):1244.
    1. Cheson BD, Fisher RI, Barrington SF, et al. ; United Kingdom National Cancer Research Institute . Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol. 2014;32(27):3059-3068.
    1. Ching T, Duncan ME, Newman-Eerkes T, et al. . Analytical evaluation of the clonoSEQ Assay for establishing measurable (minimal) residual disease in acute lymphoblastic leukemia, chronic lymphocytic leukemia, and multiple myeloma. BMC Cancer. 2020;20(1):612.
    1. Boellaard R, Delgado-Bolton R, Oyen WJ, et al. ; European Association of Nuclear Medicine (EANM) . FDG PET/CT: EANM procedure guidelines for tumour imaging: version 2.0. Eur J Nucl Med Mol Imaging. 2015;42(2):328-354.
    1. Albano D, Bosio G, Bianchetti N, et al. . Prognostic role of baseline 18F-FDG PET/CT metabolic parameters in mantle cell lymphoma. Ann Nucl Med. 2019;33(7):449-458.
    1. Bailly C, Carlier T, Berriolo-Riedinger A, et al. . Prognostic value of FDG-PET in patients with mantle cell lymphoma: results from the LyMa-PET Project. Haematologica. 2020;105(1):e33-e36.
    1. Nath K, Law SC, Sabdia MB, et al. . Intratumoral T cells have a differential impact on FDG-PET parameters in follicular lymphoma. Blood Adv. 2021;5(12):2644-2649.
    1. Schaffel R, Hedvat CV, Teruya-Feldstein J, et al. . Prognostic impact of proliferative index determined by quantitative image analysis and the International Prognostic Index in patients with mantle cell lymphoma. Ann Oncol. 2010;21(1):133-139.
    1. Saba NS, Liu D, Herman SE, et al. . Pathogenic role of B-cell receptor signaling and canonical NF-κB activation in mantle cell lymphoma. Blood. 2016;128(1):82-92.
    1. Kurtz DM, Scherer F, Jin MC, et al. . Reply to J. Wang et al. J Clin Oncol. 2019;37(9):755-757.
    1. Pittella-Silva F, Chin YM, Chan HT, et al. . Plasma or serum: which is preferable for mutation detection in liquid biopsy? Clin Chem. 2020;66(7):946-957.
    1. Rossi D, Kurtz DM, Roschewski M, Cavalli F, Zucca E, Wilson WH. The development of liquid biopsy for research and clinical practice in lymphomas: Report of the 15-ICML Workshop on ctDNA. Hematol Oncol. 2020;38(1):34-37.
    1. Epstein-Peterson ZD, Batlevi CL, Caron P, et al. . Frontline sequential immunochemotherapy plus lenalidomide for mantle cell lymphoma incorporating MRD evaluation: phase II, investigator-initiated, single-center study. Blood. 2020;136(suppl 1):11-12. Abstract 119.
    1. Scherer F, Kurtz DM, Newman AM, et al. . Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8(364):364ra155.
    1. Kurtz DM, Soo J, Co Ting Keh L, et al. . Enhanced detection of minimal residual disease by targeted sequencing of phased variants in circulating tumor DNA. Nat Biotechnol. 2021;39(12):1537-1547.

Source: PubMed

3
購読する