Improved clinical outcome in a randomized phase II study of anti-PD-1 camrelizumab plus decitabine in relapsed/refractory Hodgkin lymphoma

Yang Liu, Chunmeng Wang, Xiang Li, Liang Dong, Qingming Yang, Meixia Chen, Fengxia Shi, Malcolm Brock, Miao Liu, Qian Mei, Jiejie Liu, Jing Nie, Weidong Han, Yang Liu, Chunmeng Wang, Xiang Li, Liang Dong, Qingming Yang, Meixia Chen, Fengxia Shi, Malcolm Brock, Miao Liu, Qian Mei, Jiejie Liu, Jing Nie, Weidong Han

Abstract

Background: Programmed death-1 (PD-1) blockade monotherapy induced durable remission in a subset of patients with relapsed/refractory classical Hodgkin lymphoma (cHL). We asked whether the anti-PD-1 agent, camrelizumab, combined with the DNA demethylating agent, decitabine, improves progression-free survival (PFS) in patients with relapsed/refractory cHL over camrelizumab alone.

Methods: This extended follow-up of an ongoing randomized phase II trial analyzed PFS among patients enrolled from January 2017 through July 2018. Sixty-one patients with relapsed/refractory cHL who were clinically naïve to PD-1 blockade and had received ≥2 previous therapies were randomized 1:2 to receive either camrelizumab (200 mg) monotherapy or camrelizumab (200 mg, day 8) combined with decitabine (10 mg/day, days 1-5) every 3 weeks.

Results: With a median follow-up of 34.5 months, complete remission was 79% (95% CI 63% to 90%) in the decitabine-plus-camrelizumab group versus 32% (95% CI 13% to 57%) in the camrelizumab group (p=0.001). Median duration of response was not reached in the decitabine-plus-camrelizumab group, with an estimated 63% (95% CI 46% to 75%) of patients maintaining a response at 24 months. Median PFS with decitabine-plus-camrelizumab therapy was 35.0 months (95% CI not reached) and 15.5 months (95% CI 8.4 to 22.7 months) with camrelizumab monotherapy (HR, 0.46; 95% CI 0.21 to 1.01; p=0.02). Female gender, lower tumor burden, and fewer previous therapies were favorable prognostic factors for durable remission with camrelizumab monotherapy. The PFS benefits of decitabine-plus-camrelizumab versus camrelizumab were observed in most subgroups, especially in patients with relative larger tumor burdens and those treated with ≥3 prior therapies. After decitabine-plus-camrelizumab treatment, the percentage increase of circulating peripheral central memory T-cells correlated with both improved clinical response and PFS, suggesting a putative biomarker of decitabine-plus-camrelizumab therapy for cHL.

Conclusions: Decitabine-plus-camrelizumab results in longer PFS compared with camrelizumab alone in patients with relapsed/refractory cHL.

Trial registration numbers: NCT02961101 and NCT03250962.

Keywords: clinical trials; combination; drug therapy; immunotherapy; phase II as topic; t-lymphocytes.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Consolidated Standards of Reporting Trials diagram. CR, complete remission.
Figure 2
Figure 2
DOR in patients with CR and PR. (A) Response onset and duration for all responders. Blue bars indicate patients in the camrelizumab group and green bars indicate patients in the decitabine-plus-camrelizumab group. The length of the bar shows the time from first dosing until the patient had a CR or PR, along with the duration of the response. The onset of first response (CR or PR) was indicated as a red square (CR) or blue dot (PR), respectively. (B) Kaplan-Meier estimates of DOR in patients with CR and PR. The red curves represent patients treated with decitabine-plus-camrelizumab and the blue those treated with camrelizumab monotherapy. The median DOR and DOR rates at 12 months and 24 months are shown. Plus signs indicate censored data. (C–F) Kaplan-Meier estimates of response duration in responders who were men (C), had tumor burden SPD ≥20 cm2 (D), SPD ≥50 cm2 (E), or with ≥3 lines of previous therapy (F). The red curves represent patients treated with decitabine-plus-camrelizumab and the blue those treated with camrelizumab monotherapy. The median DOR and DOR rates at 12 months and 24 months are shown. Plus signs indicate censored data. (G) Kaplan-Meier estimates of CR duration in patients who acquired a CR after decitabine-plus-camrelizumab or camrelizumab monotherapy. The red curves represent patients treated with decitabine-plus-camrelizumab and the blue those treated with camrelizumab monotherapy. The median duration of CR was not reached and duration of CR rate at 24 months is shown. Plus signs indicate censored data. CR, complete remission; DOCR, duration of complete remission; ORR, objective response rate; PR, partial response; SPD, sum of the products of diameters.
Figure 3
Figure 3
PFS analysis. (A) Kaplan-Meier estimates of PFS in all evaluable 61 patients. The red curves represent patients treated with decitabine-plus-camrelizumab and the blue those treated with camrelizumab monotherapy. The median PFS and PFS rates at 12 months and 24 months are shown. Plus signs indicate censored data. (B) Two-year PFS rates in patient subgroups according to baseline characteristics. (C) Forest plot of median PFS for patient subgroups according to baseline demographics and disease characteristics indicating favored treatment of each subgroup. ASCT, autologous stem cell transplantation; PFS, progression-free survival; SPD, sum of the products of diameters.
Figure 4
Figure 4
Fold change of percentages in peripheral T-cell subsets during decitabine-plus-camrelizumab or camrelizumab treatment. Peripheral blood was collected from patients at the indicated times (Cnd0 indicates before the treatment cycle, n refers to treatment cycle; C1d6 indicates the day following 5-day decitabine administration in cycle 1). (A, B) Percentages of peripheral CCR7+CD45RA+ Tnaïve, CCR7+CD45RA− Tcm, CCR7−CD45RA− Tem, and CCR7−CD45RA+ Ttemra cells in CD8+ (or CD4+) T-cells at the indicated times in the camrelizumab group or decitabine-plus-camrelizumab group, analyzed by FACS. (C, D) Fold change of percentages of peripheral CCR7+CD45RA− Tcm cells in CD8+ (or CD4+) T-cells at the indicated times compared with baseline (C1d0) in the camrelizumab group or decitabine-plus-camrelizumab group, analyzed by FACS. The two-way repeated-measures analysis of variance was conducted to evaluate the effect of time–group interaction, and p value was shown. (E, F) Fold change of percentages of peripheral CCR7+CD45RA− Tcm cells in CD8+ (or CD4+) T-cells at the indicated times compared with baseline (C1d0) among patients who had different clinical responses after decitabine-plus-camrelizumab treatment. (G, H) Fold change in percentages of CCR7+CD45RA− Tcm cells in CD8+ (or CD4+) T-cells at the indicated times compared with baseline (C1d0) among patients who had different clinical responses after camrelizumab treatment. Data represent the mean±SEM. *p<0.05; **p<0.01. CR, complete response; NSHL, Nodular Sclerosis Hodgkin Lymphoma; PR, partial response; SD, stable disease.
Figure 5
Figure 5
Association of percentage alteration in peripheral T-cell subsets with CR rate and PFS. (A) The percentage of peripheral CCR7+CD45RA− Tcm, CCR7−CD45RA− Tem, CCR7+CD45RA+ Tnaïve, and CCR7−CD45RA+ Ttemra cells in CD8+ (or CD4+) T-cells detected at C5d0 or C1d6 as compared with baseline (C1d0) with CR rate in patients after camrelizumab monotherapy or decitabine-plus-camrelizumab combination. C1d6 indicates day 6 (after 5-day decitabine treatment) in the first cycle, C5d0 indicates the day before the fifth cycle. (B–E) PFS among subgroups according to CCR7+CD45RA−CD8+, CCR7+CD45RA−CD4+ ratios detected on C5d0 or C1d6 compared with baseline with decitabine-plus-camrelizumab therapy. A p value <0.05 was considered to indicate statistical significance. CR, complete response; PFS, progression-free survival.

References

    1. Hoppe RT, Advani RH, Ai WZ, et al. . Hodgkin lymphoma, version 2.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2020;18:755–81. 10.6004/jnccn.2020.0026
    1. Ansell SM, Lesokhin AM, Borrello I, et al. . Pd-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med 2015;372:311–9. 10.1056/NEJMoa1411087
    1. Younes A, Santoro A, Shipp M, et al. . Nivolumab for classical Hodgkin's lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol 2016;17:1283–94. 10.1016/S1470-2045(16)30167-X
    1. Chen R, Zinzani PL, Fanale MA, et al. . Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. J Clin Oncol 2017;35:2125–32. 10.1200/JCO.2016.72.1316
    1. Armand P, Shipp MA, Ribrag V, et al. . Programmed death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after Brentuximab Vedotin failure. J Clin Oncol 2016;34:3733–9. 10.1200/JCO.2016.67.3467
    1. Armand P, Engert A, Younes A, et al. . Nivolumab for relapsed/refractory classic Hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: extended follow-up of the Multicohort single-arm phase II CheckMate 205 trial. J Clin Oncol 2018;36:1428–39. 10.1200/JCO.2017.76.0793
    1. Armand P, Kuruvilla J, Michot J-M, et al. . KEYNOTE-013 4-year follow-up of pembrolizumab in classical Hodgkin lymphoma after brentuximab vedotin failure. Blood Adv 2020;4:12:2617–22. 10.1182/bloodadvances.2019001367
    1. Chen R, Zinzani PL, Lee HJ, et al. . Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood 2019;134:1144–53. 10.1182/blood.2019000324
    1. Shanbhag S, Ambinder RF. Hodgkin lymphoma: a review and update on recent progress. CA Cancer J Clin 2018;68:116–32. 10.3322/caac.21438
    1. Herrera AF. Where does PD-1 blockade fit in HL therapy? Hematol-Am Soc Hemat 2018;2018:213–20. 10.1182/asheducation-2018.1.213
    1. Armand P, Chen Y-B, Redd RA, et al. . Pd-1 blockade with pembrolizumab for classical Hodgkin lymphoma after autologous stem cell transplantation. Blood 2019;134:22–9. 10.1182/blood.2019000215
    1. Herrera AF, Moskowitz AJ, Bartlett NL, et al. . Interim results of brentuximab vedotin in combination with nivolumab in patients with relapsed or refractory Hodgkin lymphoma. Blood 2018;131:1183–94. 10.1182/blood-2017-10-811224
    1. Shi Y, Su H, Song Y, et al. . Safety and activity of sintilimab in patients with relapsed or refractory classical Hodgkin lymphoma (ORIENT-1): a multicentre, single-arm, phase 2 trial. Lancet Haematol 2019;6:e12–19. 10.1016/S2352-3026(18)30192-3
    1. Nie J, Wang C, Liu Y, et al. . Addition of low-dose decitabine to anti-PD-1 antibody Camrelizumab in relapsed/refractory classical Hodgkin lymphoma. J Clin Oncol 2019;37:1479–89. 10.1200/JCO.18.02151
    1. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. . Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819–29. 10.1056/NEJMoa1604958
    1. Sharma P, Hu-Lieskovan S, Wargo JA, et al. . Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. 10.1016/j.cell.2017.01.017
    1. Pitt JM, Vétizou M, Daillère R, et al. . Resistance mechanisms to Immune-Checkpoint blockade in cancer: tumor-intrinsic and -Extrinsic factors. Immunity 2016;44:1255–69. 10.1016/j.immuni.2016.06.001
    1. Ghoneim HE, Fan Y, Moustaki A, et al. . De novo epigenetic programs inhibit PD-1 Blockade-Mediated T cell rejuvenation. Cell 2017;170:e19:142–57. 10.1016/j.cell.2017.06.007
    1. Pauken KE, Sammons MA, Odorizzi PM, et al. . Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016;354:1160–5. 10.1126/science.aaf2807
    1. Wijermans P, Lübbert M, Verhoef G, et al. . Low-Dose 5-aza-2'-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase II study in elderly patients. J Clin Oncol 2000;18:956–62. 10.1200/JCO.2000.18.5.956
    1. Matei D, Fang F, Shen C, et al. . Epigenetic resensitization to platinum in ovarian cancer. Cancer Res 2012;72:2197–205. 10.1158/0008-5472.CAN-11-3909
    1. Fan H, Lu X, Wang X, et al. . Low-Dose decitabine-based chemoimmunotherapy for patients with refractory advanced solid tumors: a phase I/II report. J Immunol Res 2014;2014:1–14. 10.1155/2014/371087
    1. Biggi A, Gallamini A, Chauvie S, et al. . International validation study for interim PET in ABVD-treated, advanced-stage Hodgkin lymphoma: interpretation criteria and concordance rate among reviewers. J Nucl Med 2013;54:683–90. 10.2967/jnumed.112.110890
    1. Cheson BD, Fisher RI, Barrington SF, et al. . Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J Clin Oncol 2014;32:3059–67. 10.1200/JCO.2013.54.8800
    1. Diefenbach CS, Hong F, Ambinder RF, et al. . Ipilimumab, nivolumab, and brentuximab vedotin combination therapies in patients with relapsed or refractory Hodgkin lymphoma: phase 1 results of an open-label, multicentre, phase 1/2 trial. Lancet Haematol 2020;7:e660–70. 10.1016/S2352-3026(20)30221-0
    1. Mehta-Shah N, Bartlett NL. Management of relapsed/refractory classical Hodgkin lymphoma in transplant-ineligible patients. Blood 2018;131:1698–703. 10.1182/blood-2017-09-772681
    1. Sureda A, Constans M, Iriondo A, et al. . Prognostic factors affecting long-term outcome after stem cell transplantation in Hodgkin's lymphoma autografted after a first relapse. Ann Oncol 2005;16:625–33. 10.1093/annonc/mdi119
    1. Stiff PJ, Unger JM, Forman SJ, et al. . The value of augmented preparative regimens combined with an autologous bone marrow transplant for the management of relapsed or refractory Hodgkin disease: a southwest Oncology Group phase II trial. Biol Blood Marrow Transplant 2003;9:529–39. 10.1016/S1083-8791(03)00205-2
    1. Philip M, Fairchild L, Sun L, et al. . Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature 2017;545:452–6. 10.1038/nature22367
    1. Jansen CS, Prokhnevska N, Master VA, et al. . An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 2019;576:465–70. 10.1038/s41586-019-1836-5
    1. Miller BC, Sen DR, Al Abosy R, et al. . Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 2019;20:326–36. 10.1038/s41590-019-0312-6
    1. Weintraub K. Take two: combining immunotherapy with epigenetic drugs to tackle cancer. Nat Med 2016;22:8–10. 10.1038/nm0116-8

Source: PubMed

3
S'abonner