Daratumumab-based regimens are highly effective and well tolerated in relapsed or refractory multiple myeloma regardless of patient age: subgroup analysis of the phase 3 CASTOR and POLLUX studies

Maria-Victoria Mateos, Andrew Spencer, Ajay K Nooka, Ludek Pour, Katja Weisel, Michele Cavo, Jacob P Laubach, Gordon Cook, Shinsuke Iida, Lotfi Benboubker, Saad Z Usmani, Sung-Soo Yoon, Nizar J Bahlis, Christopher Chiu, Jon Ukropec, Jordan M Schecter, Xiang Qin, Lisa O'Rourke, Meletios A Dimopoulos, Maria-Victoria Mateos, Andrew Spencer, Ajay K Nooka, Ludek Pour, Katja Weisel, Michele Cavo, Jacob P Laubach, Gordon Cook, Shinsuke Iida, Lotfi Benboubker, Saad Z Usmani, Sung-Soo Yoon, Nizar J Bahlis, Christopher Chiu, Jon Ukropec, Jordan M Schecter, Xiang Qin, Lisa O'Rourke, Meletios A Dimopoulos

Abstract

The phase 3 POLLUX and CASTOR studies demonstrated superior benefit of daratumumab plus lenalidomide/dexamethasone or bortezomib/dexamethasone in relapsed/refractory multiple myeloma. Efficacy and safety of daratumumab was analyzed according to age groups of 65 to 74 years and ≥75 years. Patients received ≥1 prior line of therapy. In POLLUX, patients received lenalidomide/dexamethasone ± daratumumab (16 mg/kg weekly, cycles 1-2; every two weeks, cycles 3-6; monthly until progression). In CASTOR, patients received eight cycles of bortezomib/dexamethasone ± daratumumab (16 mg/kg weekly, cycles 1-3; every three weeks, cycles 4-8; monthly until progression). Patients aged >75 years received dexamethasone 20 mg weekly. For patients aged ≥75 years in POLLUX (median follow-up: 25.4 months), daratumumab/lenalido-mide/dexamethasone prolonged progression-free survival versus lenalido-mide/dexamethasone (median: 28.9 versus 11.4 months; hazard ratio, 0.27; 95% confidence interval, 0.10-0.69; P=0.0042) and increased overall response rate (93.1% versus 76.5%; P=0.0740). Neutropenia was the most common grade 3/4 treatment-emergent adverse event (daratumumab: 44.8%; control: 31.4%). Infusion-related reactions occurred in 12 (41.4%) patients. For patients aged ≥75 years in CASTOR (median follow-up: 19.4 months), daratumumab/bortezomib/dexamethasone prolonged progression-free survival versus bortezomib/dexamethasone (median: 17.9 versus 8.1 months; hazard ratio, 0.26; 95% confidence interval, 0.10-0.65; P=0.0022) and increased overall response rate (95.0% versus 78.8%; P=0.1134). Thrombocytopenia was the most common grade 3/4 treatment-emergent adverse event (daratumumab: 45.0%; control: 37.1%). Infusion-related reactions occurred in 13 (65.0%) patients. Similar findings were reported for patients aged 65 to 74 years in both studies. Taken together, this subgroup analysis of efficacy and safety of daratumumab was largely consistent with the overall populations.

Trial registration: ClinicalTrials.gov NCT02076009 NCT02136134.

Copyright© 2020 Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
Disposition of patients aged 65 to 74 years and ≥75 years based on the intent-to-treat population of (A) POLLUX and (B) CASTOR. D-Rd: daratumumab/lenalidomide/dexamethasone; Rd: lenalidomide/dexamethasone; D-Vd: daratumumab/bortezomib/dexamethasone; Vd, bortezomib/dexametha-sone. aBased on reason ‘patient refused to further study treatment’. bAll patients were to receive eight cycles of bortezomib and dexamethasone. After cycle 8, patients receiving only bortezomib and dexamethasone were entered into an observation phase, while patients in the daratumumab group continued to receive dara-tumumab monotherapy every 4 weeks. All patients had discontinued or completed eight cycles of bortezomib and dexamethasone by the interim analysis.
Figure 2.
Figure 2.
PFS of patients aged 65 to 74 years and ≥75 years in POLLUX and CASTOR. PFS in the ITT populations compared with patients aged ≥75 years (A) and 65 to 74 years (B) in POLLUX and with patients aged ≥75 years (C) and 65 to 74 years (D) in CASTOR. PFS is based on Kaplan-Meier estimates. PFS: progression-free survival; ITT: intent-to-treat; Med: median; NR: not reached; HR: hazard ratio; CI: confidence interval; Rd: lenalidomide/dexamethasone; D-Rd: daratumumab/lenalidomide/dexamethasone; Vd: bortezomib/dexamethasone; D-Vd: daratumumab/bortezomib/dexamethasone.

References

    1. Madan S, Kumar S. Current treatment options for elderly patients with multiple myeloma: clinical impact of novel agents. Therapy. 2011;8(4):415–429.
    1. Howlader N, Noone AM, Krapcho M, et al. SEER Cancer Statistics Review, 1975-2014, National Cancer Institute; 2017.
    1. Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in multiple myeloma and the impact of novel therapies. Blood. 2008;111(5):2516–2520.
    1. Palumbo A, Bringhen S, Ludwig H, et al. Personalized therapy in multiple myeloma according to patient age and vulnerability: a report of the European Myeloma Network (EMN). Blood. 2011;118(17):4519–4529.
    1. Ludwig H, Bolejack V, Crowley J, et al. Survival and years of life lost in different age cohorts of patients with multiple myeloma. J Clin Oncol. 2010;28(9):1599–1605.
    1. Willan J, Eyre TA, Sharpley F, Watson C, King AJ, Ramasamy K. Multiple myeloma in the very elderly patient: challenges and solutions. Clin Interv Aging. 2016;11:423–435.
    1. de Weers M, Tai YT, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186(3):1840–1848.
    1. Lammerts van Bueren J, Jakobs D, Kaldenhoven N, et al. Direct in vitro comparison of daratumumab with surrogate analogs of CD38 antibodies MOR03087, SAR650984 and Ab79. Blood. 2014; 124(21):3474.
    1. Overdijk MB, Verploegen S, Bogels M, et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. MAbs. 2015;7(2):311–321.
    1. Overdijk MB, Jansen JH, Nederend M, et al. The therapeutic CD38 monoclonal antibody daratumumab induces programmed cell death via Fcgamma receptor-mediated cross-linking. J Immunol. 2016;197(3):807–813.
    1. Krejcik J, Casneuf T, Nijhof IS, et al. Daratumumab depletes CD38+ immune-regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016;128(3):384–394.
    1. Chiu C, Casneuf T, Axel A, et al. Daratumumab in combination with lenalidomide plus dexamethasone induces clonality increase and T-cell expansion: results from a phase 3 randomized study (POLLUX). Blood. 2016;128(22):4531.
    1. Adams HC, III, Stevenaert F, Krejcik J, et al. High-parameter mass cytometry evaluation of relapsed/refractory multiple myeloma patients treated with daratumumab demonstrates immune modulation as a novel mechanism of action. Cytometry A. 2019;95(3):279–289.
    1. Dimopoulos MA, Oriol A, Nahi H, et al. Daratumumab, lenalidomide, and dexam-ethasone for multiple myeloma. N Engl J Med. 2016;375(14):1319–1331.
    1. Palumbo A, Chanan-Khan A, Weisel K, et al. Daratumumab, bortezomib, and dexam-ethasone for multiple myeloma. N Engl J Med. 2016;375(8):754–766.
    1. Blair HA. Daratumumab: a review in relapsed and/or refractory multiple myeloma. Drugs. 2017;77(18):2013–2024.
    1. Durie BGM, Harousseau JL, Miguel JS, et al. International uniform response criteria for multiple myeloma. Leukemia. 2006;20(9):1467–1473.
    1. Rajkumar SV, Harousseau JL, Durie B, et al. Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1. Blood. 2011;117(18): 4691–4695.
    1. Palumbo A, Bringhen S, Mateos MV, et al. Geriatric assessment predicts survival and toxicities in elderly myeloma patients: an International Myeloma Working Group report. Blood. 2015;125(13):2068–2074.
    1. Dimopoulos M, San Miguel J, Belch A, et al. Daratumumab plus lenalidomide and dex-amethasone versus lenalidomide and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of POL-LUX. Haematologica. 2018; 103(12):2088–2096.
    1. Spencer A, Lentzsch S, Weisel K, et al. Daratumumab plus bortezomib and dexamethasone versus bortezomib and dexamethasone in relapsed or refractory multiple myeloma: updated analysis of CAS-TOR. Haematologica. 2018;103(12):2079–2087.
    1. Zweegman S, Palumbo A, Bringhen S, Sonneveld P. Age and aging in blood disorders: multiple myeloma. Haematologica. 2014;99(7):1133–1137.
    1. Castelli R, Pantaleo G, Gallipoli P, et al. Salvage therapy with bortezomib and dex-amethasone in elderly patients with relapsed/refractory multiple myeloma. Anticancer Drugs. 2015;26(10):1078–1082.
    1. Dimopoulos MA, Stewart AK, Masszi T, et al. Carfilzomib, lenalidomide, and dexam-ethasone in patients with relapsed multiple myeloma categorised by age: secondary analysis from the phase 3 ASPIRE study. Br J Haematol. 2017;177(3):404–413.
    1. Dimopoulos MA, Stewart AK, Masszi T, et al. Carfilzomib-lenalidomide-dexamethasone vs lenalidomide-dexamethasone in relapsed multiple myeloma by previous treatment. Blood Cancer J. 2017;7(4):e554.
    1. Niesvizky R, Ludwig H, Spencer A, et al. Overall survival of relapsed/refractory multiple myeloma patients treated with carfilzomib and dexamethasone vs bortezomib and dexamethasone: results from the phase 3 endeavor study according to age subgroup. Presented at the 59th Annual Meeting & Exposition; December 9-12, 2017; Atlanta, GA.: 1885.
    1. San Miguel JF, Schlag R, Khuageva NK, et al. Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008;359(9):906–917.
    1. Mateos MV, Dimopoulos MA, Cavo M, et al. Daratumumab plus bortezomib, melphalan, and prednisone for untreated myeloma. N Engl J Med. 2018;378(6):518–528.
    1. Facon T, Kumar S, Plesner T, et al. Daratumumab plus lenalidomide and dexamethasone for untreated myeloma. N Engl J Med. 2019;380(22):2104–2115.
    1. Richardson PG, San Miguel JF, Moreau P, et al. Interpreting clinical trial data in multiple myeloma: translating findings to the real-world setting. Blood Cancer J. 2018;8(11):109.

Source: PubMed

3
구독하다