Daratumumab monotherapy for patients with intermediate-risk or high-risk smoldering multiple myeloma: a randomized, open-label, multicenter, phase 2 study (CENTAURUS)

C Ola Landgren, Ajai Chari, Yael C Cohen, Andrew Spencer, Peter Voorhees, Jane A Estell, Irwindeep Sandhu, Matthew W Jenner, Catherine Williams, Michele Cavo, Niels W C J van de Donk, Meral Beksac, Philippe Moreau, Hartmut Goldschmidt, Steven Kuppens, Rajesh Bandekar, Pamela L Clemens, Tobias Neff, Christoph Heuck, Ming Qi, Craig C Hofmeister, C Ola Landgren, Ajai Chari, Yael C Cohen, Andrew Spencer, Peter Voorhees, Jane A Estell, Irwindeep Sandhu, Matthew W Jenner, Catherine Williams, Michele Cavo, Niels W C J van de Donk, Meral Beksac, Philippe Moreau, Hartmut Goldschmidt, Steven Kuppens, Rajesh Bandekar, Pamela L Clemens, Tobias Neff, Christoph Heuck, Ming Qi, Craig C Hofmeister

Abstract

Current guidelines for smoldering multiple myeloma (SMM) recommend active monitoring until the onset of multiple myeloma (MM) before initiating treatment or enrollment in a clinical trial. Earlier intervention may delay progression to MM. In CENTAURUS, 123 patients with intermediate-risk or high-risk SMM were randomly assigned to daratumumab 16 mg/kg intravenously on extended intense (intense), extended intermediate (intermediate), or short dosing schedules. At the prespecified primary analysis (15.8-month median follow-up), the complete response (CR) rates (co-primary endpoint) were 2.4%, 4.9%, and 0% for intense, intermediate, and short dosing, respectively; the co-primary endpoint of CR rate >15% was not met. Progressive disease (PD)/death rates (number of patients who progressed or died divided by total duration of progression-free survival [PFS] in patient-years; co-primary endpoint) for intense, intermediate, and short dosing were 0.055 (80% confidence interval [CI], 0.014-0.096), 0.102 (80% CI, 0.044-0.160), and 0.206 (80% CI, 0.118-0.295), respectively, translating to a median PFS ≥24 months in all arms (P < 0.0001, <0.0001, and =0.0213, respectively). With longer follow-up (median follow-up, 25.9 months), CR rates were 4.9%, 9.8%, and 0% for intense, intermediate, and short dosing, respectively. PD/death rates for intense, intermediate, and short dosing were 0.059 (80% CI, 0.025-0.092), 0.107 (80% CI, 0.058-0.155), and 0.150 (80% CI, 0.089-0.211), respectively, again translating to a median PFS ≥ 24 months in all arms (P < 0.0001 for all arms). Twenty-four-month PFS rates were 89.9% (90% CI, 78.5-95.4%), 82.0% (90% CI, 69.0-89.9%), and 75.3% (90% CI, 61.1-85.0%) for intense, intermediate, and short dosing, respectively. Pharmacokinetic analyses indicated that intense dosing maintained target-saturating trough concentrations in most patients throughout weekly, every-2-week, and every-4-week dosing periods. No new safety signals were observed. These data provide the basis for an ongoing phase 3 study of daratumumab in SMM.

Trial registration: ClinicalTrials.gov NCT02316106.

Conflict of interest statement

COL received grant support from the National Institutes of Health, Food and Drug Administration, Multiple Myeloma Research Foundation, International Myeloma Foundation, Leukemia & Lymphoma Society, Perelman Family Foundation, Rising Tides Foundation, Amgen, Celgene, Janssen, Takeda, Glenmark, Seattle Genetics, and Karyopharm; received honoraria from/served on advisory committees for Adaptive Biotechnologies Corp, Amgen, Bristol-Myers Squibb, Celgene, Cellectis, Glenmark, Janssen, Karyopharm, Merck, Pfizer, Takeda, and The Binding Site; and served on independent data monitoring committees for Takeda, Novartis, and Janssen. AC received research funding from Pharmacyclics and served as a consultant for, received research funding from, and served on advisory committees for Amgen, Array BioPharma, Celgene, Janssen, Millennium, Takeda, and Novartis. YCC received research funding from Amgen and Takeda; served on advisory boards for Janssen and Takeda; served on a speakers bureau for Amgen; and received travel grants from Janssen, Bristol-Myers Squibb, and Neopharm. AS received research funding and honoraria from Janssen. PV served as a consultant for Celgene, Janssen, Bristol-Myers Squibb, Novartis, Takeda, and Oncopeptides and is on speakers bureaus for Amgen, Celgene, and Janssen. JAE served on advisory committees for Janssen and Celgene. IS received honoraria from Janssen, Celgene, Takeda, Amgen, and Novartis. MWJ received honoraria from and served on speakers bureaus for Janssen, Celgene, Takeda, Amgen, and Novartis and served on advisory committees for Janssen, Celgene, Takeda, Amgen, Novartis, and Chugai. MC received honoraria from Janssen, Celgene, Amgen, Bristol-Myers Squibb, and Takeda. NWCJvdD received research funding from Janssen, Celgene, Bristol-Myers Squibb, Novartis, and Amgen. MB served on an advisory committee and a speakers bureau for Janssen, Amgen, Celgene, and Takeda. PM served on advisory committees for Celgene, Janssen, Takeda, Novartis, Amgen, and Roche. HG consulted for and received honoraria from Celgene, Janssen-Cilag, Novartis, Bristol-Myers Squibb, Chugai, and Art Tempi; served on speakers bureaus for Adaptive Biotechnologies, Celgene, Janssen-Cilag, Bristol-Myers Squibb, Amgen, and Takeda; received compensation for travel expenses from Celgene, Bristol-Myers Squibb, Janssen-Cilag, Chugai, Amgen, and Takeda; and received research funding from Bristol-Myers Squibb, Celgene, Chugai, Janssen-Cilag, Sanofi, Novartis, Takeda, Amgen, and Mundipharma. SK, RB, TN, and CH are employees of Janssen. PLC and MQ are employees of Janssen and hold equity in Johnson & Johnson. CCH is an investigator for trials conducted by Bristol-Myers Squibb, Oncolytics Biotech, Sanofi, Celgene, Janssen, Takeda, Kite, Juno, Karyopharm, GlaxoSmithKline, Novartis, Roche, AstraZeneca, Amgen, and Aduro Biotech; received institutional research funding from Celgene, Janssen, Takeda, Roche, Karyopharm, and Bristol-Myers Squibb; served on an advisory committee for Oncopeptides, Celgene, Adaptive Biotechnologies, and Thrasos Therapeutics; and received honoraria from AXIS Medical Education, PleXus Communications, priME Oncology, TRM Oncology, DAVA Oncology, American Society of Clinical Oncology, International Myeloma Foundation, Multiple Myeloma Research Foundation, Adaptive Biotechnologies, and Thrasos Therapeutics. CW has nothing to disclose. Janssen Research & Development LLC sponsored this trial, collaborated with the academic authors on its design, compiled and maintained the data, and agreed to publish this paper.

Figures

Fig. 1. Study design and patient flow…
Fig. 1. Study design and patient flow diagram.
a Study design. b Patient flow diagram through the clinical cutoff date.aQW once weekly, Q2W every 2 weeks, Q4W every 4 weeks, Q8W every 8 weeks, IV intravenously, PD progressive disease, LPFD last patient first dose, CR complete response, MM multiple myeloma, SMM smoldering multiple myeloma, IMWG International Myeloma Working Group, FLC free light chain, PC plasma cell. aJune 29, 2018. bPD was defined per the 2014 IMWG criteria for MM [6] plus additional IMWG FLC progression criteria (a ≥25% increase from nadir in the difference between involved and uninvolved FLC levels [absolute increase must be >10 mg/dl]) [21]. cA patient could have multiple reasons for exclusion and therefore be counted in more than one category. dBone marrow PCs ≥10% to <60% plus serum M-protein ≥3 g/dl (IgA ≥2 g/dl), urine M-protein >500 mg/24 h, or abnormal FLC ratio (<0.126 or >8) and serum M-protein <3 g/dl but ≥1 g/dl. eBone marrow PCs ≥10% to <60% plus serum M-protein ≥3 g/dl (IgA ≥2 g/dl), urine M-protein >500 mg/24 h, abnormal FLC ratio (<0.126 or >8) and serum M-protein <3 g/dl but ≥1g/dl, or absolute involved serum FLC ≥100 mg/l with an abnormal FLC ratio (<0.126 or >8, but not ≤0.01 or ≥100).
Fig. 2. PFS.
Fig. 2. PFS.
PFSa with progression defined based a on diagnosticb criteria and b on biochemicalc or diagnostic criteria.dPFS progression-free survival, IMWG International Myeloma Working Group, MM multiple myeloma, FLC free light chain. aPFS was assessed in the intent-to-treat population. bDiagnostic progression was defined per the 2014 IMWG criteria for MM [6] plus additional IMWG FLC progression criteria (a ≥25% increase from nadir in the difference between involved and uninvolved FLC levels [absolute increase must be >10 mg/dl]) [21]. cBiochemical progression was defined as a measurable increase of ≥25% from nadir value in any of the following at any point during follow-up: serum M-component (absolute increase must be ≥0.5 g/dl), urine M-component (absolute increase must be ≥200 mg/24 h), and, in patients without measurable serum and urine M-protein, the difference between involved and uninvolved FLC levels (absolute increase must be >10 mg/dl). dBased on the clinical cutoff date of June 29, 2018.

References

    1. Rajkumar SV, Landgren O, Mateos MV. Smoldering multiple myeloma. Blood. 2015;125:3069–75. doi: 10.1182/blood-2014-09-568899.
    1. Landgren O, Kyle RA, Pfeiffer RM, Katzmann JA, Caporaso NE, Hayes RB, et al. Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: a prospective study. Blood. 2009;113:5412–7. doi: 10.1182/blood-2008-12-194241.
    1. Dispenzieri A, Kyle R, Katzmann JA, Therneau TM, Larson D, Benson J, et al. Immunoglobulin free light chain ratio is an independent risk factor for progression of smoldering (asymptomatic) multiple myeloma. Blood. 2008;111:785–9. doi: 10.1182/blood-2007-08-108357.
    1. Lakshman A, Rajkumar SV, Buadi FK, Binder M, Gertz MA, Lacy MQ, et al. Risk stratification of smoldering multiple myeloma incorporating revised IMWG diagnostic criteria. Blood Cancer J. 2018;8:59. doi: 10.1038/s41408-018-0077-4.
    1. Perez-Persona E, Vidriales MB, Mateo G, Garcia-Sanz R, Mateos M-V, Garcia de Coca A, et al. New criteria to identify risk of progression in monoclonal gammopathy of uncertain significance and smoldering multiple myeloma based on multiparameter flow cytometry analysis of bone marrow plasma cells. Blood. 2007;110:2586–92. doi: 10.1182/blood-2007-05-088443.
    1. Rajkumar SV, Dimopoulos MA, Palumbo A, Blade J, Merlini G, Mateos MV, et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014;15:e538–48. doi: 10.1016/S1470-2045(14)70442-5.
    1. Mateos MV, Hernandez MT, Giraldo P, de la Rubia J, de Arriba F, Lopez Corral L, et al. Lenalidomide plus dexamethasone for high-risk smoldering multiple myeloma. N Engl J Med. 2013;396:438–47. doi: 10.1056/NEJMoa1300439.
    1. Mateos MV, Hernandez MT, Giraldo P, de la Rubia J, de Arriba F, Corral LL, et al. Lenalidomide plus dexamethasone versus observation in patients with high-risk smouldering multiple myeloma (QuiRedex): long-term follow-up of a randomised, controlled, phase 3 trial. Lancet Oncol. 2016;17:1127–36. doi: 10.1016/S1470-2045(16)30124-3.
    1. Lin P, Owens R, Tricot G, Wilson CS. Flow cytometric immunophenotypic analysis of 306 cases of multiple myeloma. Am J Clin Pathol. 2004;121:482–8. doi: 10.1309/74R4TB90BUWH27JX.
    1. Santonocito AM, Consoli U, Bagnato S, Milone G, Palumbo GA, Di Raimondo F, et al. Flow cytometric detection of aneuploid CD38(++) plasmacells and CD19(+) B-lymphocytes in bone marrow, peripheral blood and PBSC harvest in multiple myeloma patients. Leuk Res. 2004;28:469–77. doi: 10.1016/j.leukres.2003.09.015.
    1. de Weers M, Tai YT, van der Veer MS, Bakker JM, Vink T, Jacobs DC, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol. 2011;186:1840–8. doi: 10.4049/jimmunol.1003032.
    1. Lammerts van Bueren J, Jakobs D, Kaldenhoven N, Roza M, Hiddingh S, Meesters J, et al. Direct in vitro comparison of daratumumab with surrogate analogs of CD38 antibodies MOR03087, SAR650984 and Ab79. Blood. 2014;124:3474. doi: 10.1182/blood.V124.21.3474.3474.
    1. Overdijk MB, Verploegen S, Bogels M, van Egmond M, Lammerts van Bueren JJ, Mutis T, et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. MAbs. 2015;7:311–21. doi: 10.1080/19420862.2015.1007813.
    1. van de Donk NWCJ, Janmaat ML, Mutis T, Lammerts van Bueren JJ, Ahmadi T, Sasser AK, et al. Monoclonal antibodies targeting CD38 in hematological malignancies and beyond. Immunol Rev. 2016;270:95–112. doi: 10.1111/imr.12389.
    1. Krejcik J, Casneuf T, Nijhof IS, Verbist B, Bald J, Plesner T, et al. Daratumumab depletes CD38+ immune-regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016;128:384–94. doi: 10.1182/blood-2015-12-687749.
    1. Overdijk MB, Jansen JH, Nederend M, Lammerts van Bueren JJ, Groen RW, Parren PW, et al. The therapeutic CD38 monoclonal antibody daratumumab induces programmed cell death via Fcgamma receptor-mediated cross-linking. J Immunol. 2016;197:807–13. doi: 10.4049/jimmunol.1501351.
    1. Lokhorst HM, Plesner T, Laubach JP, Nahi H, Gimsing P, Hansson M, et al. Targeting CD38 with daratumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373:1207–19. doi: 10.1056/NEJMoa1506348.
    1. Lonial S, Weiss BM, Usmani S, Singhal S, Chari A, Bahlis N, et al. Daratumumab monotherapy in patients with treatment-refractory multiple myeloma (SIRIUS): an open-label, randomised, phase 2 trial. Lancet. 2016;387:1551–60. doi: 10.1016/S0140-6736(15)01120-4.
    1. Brighton TA, Khot A, Harrison SJ, Ghez D, Weiss BM, Kirsch A, et al. Randomized, double-blind, placebo-controlled, multicenter study of siltuximab in high-risk smoldering multiple myeloma. Clin Cancer Res. 2019;25:3772–5. doi: 10.1158/1078-0432.CCR-18-3470.
    1. Dispenzieri A, Stewart AK, Chanan-Khan A, Rajkumar SV, Kyle RA, Fonseca R, et al. Smoldering multiple myeloma requiring treatment: time for a new definition? Blood. 2013;122:4172–81. doi: 10.1182/blood-2013-08-520890.
    1. Durie BGM, Harousseau JL, Miguel JS, Blade J, Barlogie B, Anderson K, et al. International uniform response criteria for multiple myeloma. Leukemia. 2006;20:1467–73. doi: 10.1038/sj.leu.2404284.
    1. Rajkumar SV, Harousseau JL, Durie B, Anderson KC, Dimopoulos M, Kyle R, et al. Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1. Blood. 2011;117:4691–5. doi: 10.1182/blood-2010-10-299487.
    1. McCudden C, Axel AE, Slaets D, Dejoie T, Clemens PL, Frans S, et al. Monitoring multiple myeloma patients treated with daratumumab: teasing out monoclonal antibody interference. Clin Chem Lab Med. 2016;54:1095–104. doi: 10.1515/cclm-2015-1031.
    1. US Department of Health and Human Services, National Institutes of Health, National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE). Version 4.03. . Accessed 4 Jan 2019.
    1. Ghosh A, Mailankody S, Giralt SA, Landgren CO, Smith EL, Brentjens RJ. CAR T cell therapy for multiple myeloma: where are we now and where are we headed? Leuk Lymphoma. 2018;59:2056–67. doi: 10.1080/10428194.2017.1393668.
    1. Ravi P, Kumar S, Larsen JT, Gonsalves W, Buadi F, Lacy MQ, et al. Evolving changes in disease biomarkers and risk of early progression in smoldering multiple myeloma. Blood Cancer J. 2016;6:e454. doi: 10.1038/bcj.2016.65.
    1. Fernandez de Larrea C, Isola I, Pereira A, Cibeira MT, Magnano L, Tovar N, et al. Evolving M-protein pattern in patients with smoldering multiple myeloma: impact on early progression. Leukemia. 2018;32:1427–34. doi: 10.1038/s41375-018-0013-4.
    1. Wu V, Moshier E, Leng S, Barlogie B, Cho HJ, Jagannath S, et al. Risk stratification of smoldering multiple myeloma: predictive value of free light chains and group-based trajectory modeling. Blood Adv. 2018;2:1470–9. doi: 10.1182/bloodadvances.2018016998.
    1. Landgren O. Shall we treat smoldering multiple myeloma in the near future? Hematol Am Soc Hematol Educ Program. 2017;2017:194–204. doi: 10.1182/asheducation-2017.1.194.
    1. Korde N, Roschewski M, Zingone A, Kwok M, Manasanch EE, Bhutani M, et al. Treatment with carfilzomib-lenalidomide-dexamethasone with lenalidomide extension in patients with smoldering or newly diagnosed multiple myeloma. JAMA Oncol. 2015;1:746–54. doi: 10.1001/jamaoncol.2015.2010.
    1. Mateos MV, Martinez Lopez J, Rodriguez-Otero P, Ocio EM, Gonzalez MS, Oriol A, et al. Curative strategy for high-risk smoldering myeloma (GEM-CESAR): carfilzomib, lenalidomide and dexamethasone (KRd) as induction followed by HDT-ASCT, consolidation with Krd and maintenance with Rd. Blood. 2017;130:402.
    1. Lonial S, Jacobus SJ, Weiss M, Kumar S, Orlowski RZ, Kaufman JL, et al. E3A06: Randomized phase III trial of lenalidomide versus observation alone in patients with asymptomatic high-risk smoldering multiple myeloma. J Clin Oncol. 2019;37:8001. doi: 10.1200/JCO.2019.37.15_suppl.8001.
    1. Xu XS, Yan X, Puchalski T, Lonial S, Lokhorst HM, Voorhees PM, et al. Clinical implications of complex pharmacokinetics for daratumumab dose regimen in patients with relapsed/refractory multiple myeloma. Clin Pharm Ther. 2017;101:721–4. doi: 10.1002/cpt.577.
    1. Clemens PL, Yan X, Lokhorst HM, Lonial S, Losic N, Khan I, et al. Pharmacokinetics of daratumumab following intravenous infusion in relapsed or refractory multiple myeloma after prior proteasome inhibitor and immunomodulatory drug treatment. Clin Pharmacokinet. 2016;56:915–24. doi: 10.1007/s40262-016-0477-1.
    1. Yan X, Clemens PL, Puchalski T, Lonial S, Lokhorst HM, Orlowski RZ, et al. Target-mediated drug disposition of daratumumab following intravenous infusion in relapsed or refractory multiple myeloma after prior proteasome inhibitors and immunomodulatory drugs: a population pharmacokinetic analysis. Blood. 2015;126:4222. doi: 10.1182/blood.V126.23.4222.4222.

Source: PubMed

3
Tilaa