512 results on '"T. Facon"'
Search Results
152. COVID-19 vaccination in patients with multiple myeloma: a consensus of the European Myeloma Network.
- Author
-
Ludwig H, Sonneveld P, Facon T, San-Miguel J, Avet-Loiseau H, Mohty M, Mateos MV, Moreau P, Cavo M, Pawlyn C, Zweegman S, Engelhardt M, Driessen C, Cook G, Dimopoulos MA, Gay F, Einsele H, Delforge M, Caers J, Weisel K, Jackson G, Garderet L, van de Donk N, Leleu X, Goldschmidt H, Beksac M, Nijhof I, Schreder M, Abildgaard N, Hajek R, Zojer N, Kastritis E, Broijl A, Schjesvold F, Boccadoro M, and Terpos E
- Subjects
- Consensus, Humans, Multiple Myeloma drug therapy, Multiple Myeloma immunology, SARS-CoV-2, Vaccination, COVID-19 prevention & control, COVID-19 Vaccines administration & dosage, Multiple Myeloma complications, Practice Guidelines as Topic standards
- Abstract
Patients with multiple myeloma frequently present with substantial immune impairment and an increased risk for infections and infection-related mortality. The risk for infection with SARS-CoV-2 virus and resulting mortality is also increased, emphasising the importance of protecting patients by vaccination. Available data in patients with multiple myeloma suggest a suboptimal anti-SARS-CoV-2 immune response, meaning a proportion of patients are unprotected. Factors associated with poor response are uncontrolled disease, immunosuppression, concomitant therapy, more lines of therapy, and CD38 antibody-directed and B-cell maturation antigen-directed therapy. These facts suggest that monitoring the immune response to vaccination in patients with multiple myeloma might provide guidance for clinical management, such as administration of additional doses of the same or another vaccine, or even temporary treatment discontinuation, if possible. In those who do not exhibit a good response, prophylactic treatment with neutralising monoclonal antibody cocktails might be considered. In patients deficient of a SARS-CoV-2 immune response, adherence to measures for infection risk reduction is particularly recommended. This consensus was generated by members of the European Multiple Myeloma Network and some external experts. The panel members convened in virtual meetings and conducted an extensive literature research and evaluated recently published data and work presented at meetings, as well as findings from their own studies. The outcome of the discussions on establishing consensus recommendations for COVID-19 vaccination in patients with multiple myeloma was condensed into this Review., Competing Interests: Declaration of interests HL declares research funding from Amgen and Takeda, and speaker's honoraria from and participation on advisory boards for Amgen, Takeda, Sanofi, Janssen, Celgene-Bristol Myers Squibb, and Seattle Genetics. PS declares research funding from Amgen, Celgene-Bristol Myers Squibb, Janssen, SkylineDx, and Takeda, and honoraria from and participation on advisory boards for Amgen, Celgene-Bristol Myers Squibb, Janssen, SkylineDx, and Takeda. TF declares participation on advisory boards for Janssen, Bristol Myers Squibb, Takeda, Amgen, Roche, Karyopharm, Oncopeptides, and Abbvie, and speaker's honoraria from Janssen and Bristol Myers Squibb. JS-M declares consulting fees from and participation on advisory boards for Amgen, Celgene-Bristol Myers Squibb, Janssen, MSD, Novartis, Takeda, Sanofi, Roche, Abbvie, GlaxoSmithKline, Regeneron, SecuraBio, and Karyopharm. M-VM declares honoraria from and participation on advisory boards for Janssen, Celgene-Bristol Myers Squibb, Takeda, Amgen, Sanofi, Oncopeptides, GlaxoSmithKline, Adaptive, Pfizer, Regeneron, Roche, Sea-Gen, and Blu Bird bio. PM declares honoraria from and participation on advisory boards for Janssen, Celgene-Bristol Myers Squibb, Amgen, Sanofi, and Abbvie. MC declares honoraria from Janssen, Celgene-Bristol Myers Squibb, GlaxoSmithKline, Amgen, Takeda, AbbVie, and Sanofi, and participation on advisory boards for Janssen, Celgene-Bristol Myers Squibb, GlaxoSmithKline, Amgen, Takeda, AbbVie, and Sanofi. CP declares consultancy fees from Amgen, Takeda, Celgene-Bristol Myers Squibb, and Sanofi; travel support from Amgen, Takeda, Janssen, and Celgene-Bristol Myers Squibb; and honoraria from Janssen, Celgene-Bristol Myers Squibb, and Sanofi. SZ declares research funding from Takeda and Janssen, and participation on advisory boards for Takeda, Janssen, Sanofi, Bristol Myers Squibb, and Oncopeptides. ME declares honoraria from and participation on advisory boards for Amgen, Celgene-Bristol Myers Squibb, GlaxoSmithKline, Janssen, Karyopharm, Sanofi, and Takeda, and research funding from Amgen, Celgene-Bristol Myers Squibb, Janssen, Karyopharm, and Takeda. GC declares honoraria from and participation on advisory boards for Celgene-Bristol Myers Squibb, Takeda, GlaxoSmithKline, Sanofi, Amgen, Janssen, and Oncopeptides, and research funding from GlaxoSmithKline and Takeda. MAD declares participation on advisory boards for Amgen, Takeda, Bristol Myers Squibb, Janssen, and Beigene. FG declares honoraria from Amgen, Janssen, Takeda, Celgene-Bristol Myers Squibb, AbbVie, and GlaxoSmithKline, and participation on advisory boards for Amgen, Celgene-Bristol Myers Squibb, Janssen, Takeda, AbbVie, GlaxoSmithKline, Roche, Adaptive Biotechnologies, Oncopeptides, and Bluebird Bio. HE declares honoraria from Amgen, Celgene-Bristol Myers Squibb, Janssen, Takeda, GlaxoSmithKline, Sanofi, Novartis; consultancy fees from and participation on advisory boards for Amgen, Celgene-Bristol Myers Squibb, Janssen, Takeda, GlaxoSmithKline, Sanofi, and Novartis; and research funding from Amgen, Celgene-Bristol Myers Squibb, Janssen, Sanofi, and GlaxoSmithKline. MD declares speaker's honoraria and research funding from Amgen, Celgene-Bristol Myers Squibb, Janssen, Sanofi, and Takeda. KW declares research funding from Amgen, Celgene-Bristol Myers Squibb, Janssen, and Sanofi, and honoraria from Amgen, Abbvie, Adaptive Biotech, Celgene-Bristol Myers Squibb, Janssen, Karyopharm, Novartis, Oncopeptides, Roche Pharma, Takeda, and Sanofi. GJ declares speaker's honoraria from and participation on advisory boards for Celgene-Bristol Myers Squibb, Takeda, GlaxoSmithKline, Sanofi, Amgen, Johnson & Johnson, and Oncopeptides, and research funding from Celgene-Bristol Myers Squibb and Takeda. LG declares participation on advisory boards for Amgen, Takeda, Celgene-Bristol Myers Squibb, and Janssen. NvdD declares research funding from Janssen Pharmaceuticals, Amgen, Celgene-Bristol Myers Squibb, Novartis, and Cellectis, and participation on advisory boards for Janssen Pharmaceuticals, Amgen, Celgene-Bristol Myers Squibb, Takeda, Roche, Novartis, Bayer, Servier, GlaxoSmithKline, and Sanofi. XL declares honoraria from Janssen-Cilag, Celgene-Bristol Myers Squibb, Amgen, Novartis, Takeda, Sanofi, Abbvie, Merck, Roche, Karyopharm Therapeutics, Carsgen Therapeutics, Oncopeptides, and GlaxoSmithKline; consulting and advisory roles for Janssen-Cilag, Celgene-Bristol Myers Squibb, Amgen, Takeda, Novartis, Merck, Gilead Sciences, Abbvie, Roche, Karyopharm Therapeutics, Oncopeptides, Carsgen Therapeutics, and GlaxoSmithKline; travel fees from Accommodations; and expenses from Takeda. HG declares grants and provision of Investigational Medicinal Product Amgen, Celgene-Bristol Myers Squibb, Chugai, Janssen, and Sanofi; research support from Amgen, Celgene-Bristol Myers Squibb, Chugai, Janssen, Incyte, Molecular Partners, MSD, Sanofi, Mundipharma GmbH, Takeda, and Novartis; participation on advisory boards for Adaptive Biotechnology, Amgen, Celgene-Bristol Myers Squibb, Janssen, Sanofi, and Takeda; and honoraria from Amgen, Celgene-Bristol Myers Squibb, Chugai, GlaxoSmithKline, Janssen, Novartis, and Sanofi. MeB declares participation on advisory boards for Amgen, Celgene-Bristol Myers Squibb, Janssen, Sanofi, Takeda, and Oncopeptides, and speaker's honoraria from Amgen, Celgene-Bristol Myers Squibb, Janssen, Sanofi, and Takeda. IN declares Advisory Boards and Honoraria: Amgen, Janssen, Celgene-Bristol Myers Squibb. MS declares speaker's honoraria and participation on advisory boards for Celgene-Bristol Myers Squibb, Amgen, Takeda, Janssen, and GlaxoSmithKline. NA declares research funding from Celgene-Bristol Myers Squibb, Amgen, Janssen, and Takeda. RH declares consultancy fees from and participation on advisory boards for Janssen, Amgen, AbbVie, Bristol Myers Squibb, Novartis, PharmaMar, and Takeda; honoraria from Janssen, Amgen, Bristol Myers Squibb, PharmaMar, and Takeda; and research funding from Janssen, Amgen, Bristol Myers Squibb, Novartis, and Takeda. NZ declares speaker's honoraria and participation on advisory boards for Celgene-Bristol Myers Squibb, Amgen, Takeda, Janssen, and Sanofi. EK declares consultancy fees and honoraria from and participation on advisory boards for Amgen, Genesis Pharma, Takeda, Janssen, and Pfizer Sanofi. AB declares honoraria from and participation on advisory boards for Amgen, Janssen, Celgene-Bristol Myers Squibb, and Sanofi. FS declares honoraria from Amgen, Celgene, Janssen, MSD, Novartis, Oncopeptides, Sanofi, SkyliteDX, and Takeda, and membership on an entity's advisory committees for Amgen, Celgene-Bristol Myers Squibb, Janssen, MSD, Novartis, Oncopeptides, Sanofi, and Takeda. MaB declares honoraria from Sanofi, Celgene-Bristol Myers Squibb, Amgen, Janssen, Novartis, and AbbVie; participation on advisory boards for Janssen and GlaxoSmithKline; and research funding from Sanofi, Celgene-Bristol Myers Squibb, Amgen, Janssen, Novartis, and Mundipharma. ET declares consultancy fees and honoraria from Amgen, Celgene-Bristol Myers Squibb, Janssen, Takeda, Genesis Pharma, GlaxoSmithKline, and Sanofi, and research support from Amgen, Janssen, Celgene-Bristol Myers Squibb, Genesis Pharma, GlaxoSmithKline, and Sanofi. HA-L, MM, CD, and JC declare no competing interests., (Crown Copyright © 2021 Published by Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
153. Isatuximab plus pomalidomide and dexamethasone in frail patients with relapsed/refractory multiple myeloma: ICARIA-MM subgroup analysis.
- Author
-
Schjesvold F, Bringhen S, G Richardson P, Perrot A, Leleu X, Moreau P, A Dimopoulos M, Hulin C, Tekle C, Foster MC, Poole EM, van de Velde H, and Facon T
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Agents, Immunological therapeutic use, Frail Elderly, Humans, Thalidomide therapeutic use, Treatment Outcome, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Thalidomide analogs & derivatives
- Published
- 2021
- Full Text
- View/download PDF
154. Daratumumab, lenalidomide, and dexamethasone versus lenalidomide and dexamethasone alone in newly diagnosed multiple myeloma (MAIA): overall survival results from a randomised, open-label, phase 3 trial.
- Author
-
Facon T, Kumar SK, Plesner T, Orlowski RZ, Moreau P, Bahlis N, Basu S, Nahi H, Hulin C, Quach H, Goldschmidt H, O'Dwyer M, Perrot A, Venner CP, Weisel K, Mace JR, Raje N, Tiab M, Macro M, Frenzel L, Leleu X, Ahmadi T, Wang J, Van Rampelbergh R, Uhlar CM, Tromp B, Delioukina M, Vermeulen J, and Usmani SZ
- Subjects
- Aged, Drug-Related Side Effects and Adverse Reactions diagnosis, Drug-Related Side Effects and Adverse Reactions epidemiology, Female, Humans, Male, Progression-Free Survival, Survival Rate, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Multiple Myeloma mortality
- Abstract
Background: In the primary analysis of the phase 3 MAIA trial (median follow-up 28·0 months), a significant improvement in progression-free survival was observed with daratumumab plus lenalidomide and dexamethasone versus lenalidomide and dexamethasone alone in transplantation-ineligible patients with newly diagnosed multiple myeloma. Here, we report the updated efficacy and safety results from a prespecified interim analysis for overall survival., Methods: MAIA is an ongoing, multicentre, randomised, open-label, phase 3 trial that enrolled patients at 176 hospitals in 14 countries across North America, Europe, the Middle East, and the Asia-Pacific region. Eligible patients were aged 18 years or older, had newly diagnosed multiple myeloma, had an Eastern Cooperative Oncology Group performance status score of 0-2, and were ineligible for high-dose chemotherapy with autologous stem-cell transplantation because of their age (≥65 years) or comorbidities. Patients were randomly assigned (1:1) using randomly permuted blocks (block size 4) by an interactive web response system to receive 28-day cycles of intravenous daratumumab (16 mg/kg, once per week during cycles 1-2, once every 2 weeks in cycles 3-6, and once every 4 weeks thereafter) plus oral lenalidomide (25 mg on days 1-21 of each cycle) and oral dexamethasone (40 mg on days 1, 8, 15, and 22 of each cycle; daratumumab group) or lenalidomide and dexamethasone alone (control group). Randomisation was stratified by International Staging System disease stage, geographical region, and age. Neither patients nor investigators were masked to treatment assignment. The primary endpoint was progression-free survival, which was centrally assessed, and a secondary endpoint was overall survival (both assessed in the intention-to-treat population). The safety population included patients who received at least one dose of the study treatment. The results presented here are from a prespecified interim analysis for overall survival, for which the prespecified stopping boundary was p=0·0414. This trial is registered with ClinicalTrials.gov, NCT02252172., Findings: Between March 18, 2015, and Jan 15, 2017, 952 patients were assessed for eligibility, of whom 737 patients were enrolled and randomly assigned to the daratumumab group (n=368) or the control group (n=369). At a median follow-up of 56·2 months (IQR 52·7-59·9), median progression-free survival was not reached (95% CI 54·8-not reached) in the daratumumab group versus 34·4 months (29·6-39·2) in the control group (hazard ratio [HR] 0·53 [95% CI 0·43-0·66]; p<0·0001). Median overall survival was not reached in either group (daratumumab group, 95% CI not reached-not reached; control group, 95% CI 55·7-not reached; HR 0·68 [95% CI 0·53-0·86]; p=0·0013). The most common (>15%) grade 3 or higher treatment-emergent adverse events were neutropenia (197 [54%] patients in the daratumumab group vs 135 [37%] patients in the control group), pneumonia (70 [19%] vs 39 [11%]), anaemia (61 [17%] vs 79 [22%]), and lymphopenia (60 [16%] vs 41 [11%]). Serious adverse events occurred in 281 (77%) patients in the daratumumab group and 257 (70%) patients in the control group. Treatment-related deaths occurred in 13 (4%) patients in the daratumumab group and ten (3%) patients in the control group., Interpretation: Daratumumab plus lenalidomide and dexamethasone increased overall survival and progression-free survival in patients ineligible for stem-cell transplantation with newly diagnosed multiple myeloma. There were no new safety concerns. Our results support the frontline use of daratumumab plus lenalidomide and dexamethasone for patients with multiple myeloma who are ineligible for transplantation., Funding: Janssen Research & Development., Competing Interests: Declaration of interests TF received payment or honoraria for speakers bureaus for Bristol Myers Squibb, Janssen, and Takeda; and participated on advisory boards for AbbVie, Amgen, Bristol Myers Squibb, Janssen, Karyopharm, Oncopeptides, Roche, and Takeda. SKK received research funding from AbbVie, Amgen, AstraZeneca, Bristol Myers Squibb, CARsgen, Janssen, Molecular Templates, Novartis, Roche-Genentech, Takeda, and TeneoBio; and consulted for or participated on an advisory board for AbbVie, Amgen, Antengene, AstraZeneca, BeiGene, bluebird bio, Bristol Myers Squibb, Epizyme, Janssen, Oncopeptides, Roche-Genentech, Secure Biotherapeutics, and Takeda. TP received consulting fees from Celgene and Janssen; received payment or honoraria for educational events from Janssen; received payment for expert testimony from CSL Behring, Oncopeptides, and Takeda; and holds stock or stock options in Novo Nordisk. RZO received support for the present manuscript from Janssen; received laboratory research funding from Asylia Therapeutics, BioTheryX, and Heidelberg Pharma; received clinical research funding from CARsgen Therapeutics, Celgene, Exelixis, Janssen Biotech, Sanofi-Aventis, and Takeda; received royalties or licenses for, has patents planned, issued or pending for, and holds stock or stock options in Asylia Therapeutics; received consulting fees from EcoR1 Capital; and participated on an advisory board for Amgen, BioTheryX, Bristol Myers Squibb, Celgene, Forma Therapeutics, Genzyme, GSK Biologicals, Ionis, Janssen Biotech, Juno Therapeutics, Karyopharm, Kite, Neoleukin, Oncopeptides, Regeneron, Sanofi-Aventis, Servier, and Takeda. PM received honoraria from AbbVie, Amgen, Celgene, Janssen, Oncopeptides, and Sanofi. NB received research funding from Celgene, Janssen, and Pfizer; participated on an independent review committee for Janssen, Karyopharm, and Legend Biotech; received honoraria for educational events from Bristol Myers Squibb, Celgene, and Janssen; and participated on an advisory board for AbbVie, Amgen, Bristol Myers Squibb, Celgene, Genentech, GlaxoSmithKline, Janssen, Karyopharm, Pfizer, Sanofi, and Takeda. HQ received grants or contracts from Amgen, Celgene, GlaxoSmithKline, Karyopharm, and Sanofi; and had a leadership or fiduciary role in advisory boards for Amgen, Celgene, CSL Behring, GlaxoSmithKline, Janssen, Karyopharm, Sanofi, and Takeda. HG received grants or contracts from Amgen, Bristol Myers Squibb, Celgene, Chugai, Dietmar-Hopp-Foundation, Janssen, Johns Hopkins University, and Sanofi; received consulting fees from Adaptive Biotechnologies, Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi, and Takeda; received payment or honoraria for lectures and presentations from Amgen, Art Tempi, Bristol Myers Squibb, Celgene, Chugai, GlaxoSmithKline, Janssen, Novartis, and Sanofi; received support for attending meetings or travel, or both, from Adaptive Biotechnologies, Amgen, Art Tempi, Bristol Myers Squibb, Celgene, Chugai, Janssen, Sanofi, and Takeda; and received research support from Amgen, Bristol Myers Squibb, Celgene, Chugai, Incyte, Janssen, Merck Sharp & Dohme, Molecular Partners, MundiPharma, Novartis, Sanofi, and Takeda. MOD received consulting fees from Janssen; has several patents planned, issued, or pending relating to the use of natural killer cells and sialyltransferase inhibition in the treatment of cancer; participated on an advisory board for Amgen, Celgene, and Janssen; serves as a director of ONK Therapeutics; and holds stock or stock options in ONK Therapeutics and Carrick Therapeutics. AP received grants or contracts from Sanofi and Takeda; received payment or honoraria for presentations and educational events from AbbVie, Amgen, Bristol Myers Squibb/Celgene, GlaxoSmithKline, Janssen, Sanofi, and Takeda; received payment or honoraria for a speakers bureau from Janssen and Sanofi; received support for attending meetings or travel, or both, from Amgen and Janssen; and participated on a data safety monitoring board or advisory board for Janssen and Sanofi. CPV received honoraria from Amgen, Bristol Myers Squibb, GlaxoSmithKline, Janssen, Pfizer, Sanofi, and Takeda. KW received honoraria from AbbVie, Adaptive Biotechnologies, Amgen, Bristol Myers Squibb/Celgene, GlaxoSmithKline, Janssen, Karyopharm, Novartis, Oncopeptides, Pfizer, Roche, Sanofi, and Takeda; and received grants from Amgen, Bristol Myers Squibb/Celgene, GlaxoSmithKline, and Sanofi. NR served in an advisory role for Amgen, bluebird bio, Caribou, GlaxoSmithKline, and Immuneel. MM received research funding from Janssen and Takeda; received honoraria from GlaxoSmithKline, Janssen, Sanofi, and Takeda; and received travel accommodations from Janssen and Takeda. LF received support for attending meetings or travel, or both, from Amgen and Janssen; and received payment or honoraria for educational events from Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi, and Takeda. TA is an employee of and holds equity in Genmab. JW and RVR are employees of Janssen. CMU, BT, MD, and JV are employees of Janssen; and hold equity in Johnson & Johnson. SZU received grants from Amgen, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Merck, Pharmacyclics, Sanofi, Seattle Genetics, SkylineDX, and Takeda; and received personal fees from AbbVie, Amgen, Celgene, Genentech, Gilead, GlaxoSmithKline, Janssen, Merck, MundiPharma, Oncopeptides, Sanofi, Seattle Genetics, SkylineDX, and Takeda. SB, HN, CH, JRM, MT, and XL declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
155. Minimal Residual Disease in Myeloma: Application for Clinical Care and New Drug Registration.
- Author
-
Anderson KC, Auclair D, Adam SJ, Agarwal A, Anderson M, Avet-Loiseau H, Bustoros M, Chapman J, Connors DE, Dash A, Di Bacco A, Du L, Facon T, Flores-Montero J, Gay F, Ghobrial IM, Gormley NJ, Gupta I, Higley H, Hillengass J, Kanapuru B, Kazandjian D, Kelloff GJ, Kirsch IR, Kremer B, Landgren O, Lightbody E, Lomas OC, Lonial S, Mateos MV, Montes de Oca R, Mukundan L, Munshi NC, O'Donnell EK, Orfao A, Paiva B, Patel R, Pugh TJ, Ramasamy K, Ray J, Roshal M, Ross JA, Sigman CC, Thoren KL, Trudel S, Ulaner G, Valente N, Weiss BM, Zamagni E, and Kumar SK
- Subjects
- Bone Marrow, High-Throughput Nucleotide Sequencing methods, Humans, Neoplasm, Residual diagnosis, Retrospective Studies, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy
- Abstract
The development of novel agents has transformed the treatment paradigm for multiple myeloma, with minimal residual disease (MRD) negativity now achievable across the entire disease spectrum. Bone marrow-based technologies to assess MRD, including approaches using next-generation flow and next-generation sequencing, have provided real-time clinical tools for the sensitive detection and monitoring of MRD in patients with multiple myeloma. Complementary liquid biopsy-based assays are now quickly progressing with some, such as mass spectrometry methods, being very close to clinical use, while others utilizing nucleic acid-based technologies are still developing and will prove important to further our understanding of the biology of MRD. On the regulatory front, multiple retrospective individual patient and clinical trial level meta-analyses have already shown and will continue to assess the potential of MRD as a surrogate for patient outcome. Given all this progress, it is not surprising that a number of clinicians are now considering using MRD to inform real-world clinical care of patients across the spectrum from smoldering myeloma to relapsed refractory multiple myeloma, with each disease setting presenting key challenges and questions that will need to be addressed through clinical trials. The pace of advances in targeted and immune therapies in multiple myeloma is unprecedented, and novel MRD-driven biomarker strategies are essential to accelerate innovative clinical trials leading to regulatory approval of novel treatments and continued improvement in patient outcomes., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
156. Maintenance with daratumumab or observation following treatment with bortezomib, thalidomide, and dexamethasone with or without daratumumab and autologous stem-cell transplant in patients with newly diagnosed multiple myeloma (CASSIOPEIA): an open-label, randomised, phase 3 trial.
- Author
-
Moreau P, Hulin C, Perrot A, Arnulf B, Belhadj K, Benboubker L, Béné MC, Zweegman S, Caillon H, Caillot D, Corre J, Delforge M, Dejoie T, Doyen C, Facon T, Sonntag C, Fontan J, Mohty M, Jie KS, Karlin L, Kuhnowski F, Lambert J, Leleu X, Macro M, Orsini-Piocelle F, Roussel M, Stoppa AM, van de Donk NWCJ, Wuillème S, Broijl A, Touzeau C, Tiab M, Marolleau JP, Meuleman N, Vekemans MC, Westerman M, Klein SK, Levin MD, Offner F, Escoffre-Barbe M, Eveillard JR, Garidi R, Ahmadi T, Krevvata M, Zhang K, de Boer C, Vara S, Kampfenkel T, Vanquickelberghe V, Vermeulen J, Avet-Loiseau H, and Sonneveld P
- Subjects
- Adolescent, Adult, Aged, Antibodies, Monoclonal adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib adverse effects, Dexamethasone adverse effects, Drug Administration Schedule, Europe, Female, Humans, Maintenance Chemotherapy, Male, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma mortality, Progression-Free Survival, Thalidomide adverse effects, Time Factors, Transplantation, Autologous, Young Adult, Antibodies, Monoclonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Bortezomib administration & dosage, Dexamethasone administration & dosage, Multiple Myeloma therapy, Stem Cell Transplantation adverse effects, Thalidomide administration & dosage
- Abstract
Background: CASSIOPEIA part 1 showed superior depth of response and significantly improved progression-free survival with daratumumab, bortezomib, thalidomide, and dexamethasone (D-VTd) versus bortezomib, thalidomide, and dexamethasone (VTd) as induction and consolidation in patients with autologous stem-cell transplant (ASCT)-eligible newly diagnosed multiple myeloma. In part 2, we compared daratumumab maintenance versus observation only., Methods: CASSIOPEIA is a two-part, open-label, randomised, phase 3 trial of patients aged 18-65 years with newly diagnosed multiple myeloma and Eastern Cooperative Oncology Group performance status 0-2, done in 111 European academic and community practice centres. In part 1, patients were randomly assigned (1:1) to induction and consolidation with D-VTd or VTd. Patients still on study who had a partial response or better were randomly assigned (1:1) by an interactive web-response system to daratumumab 16 mg/kg intravenously every 8 weeks (a reduced frequency compared with standard daratumumab long-term dosing) or observation only for up to 2 years. Stratification factors were induction treatment and depth of response in part 1. The part 2 primary endpoint was progression-free survival from second randomisation. This preplanned interim analysis of progression-free survival was done after 281 events and shall be considered the primary analysis of progression-free survival. Sponsor personnel and designees who were involved in the analysis were masked to treatment group until the independent data monitoring committee recommended that the preplanned interim analysis be considered the main analysis of progression-free survival in part 2. Otherwise, treatment assignments were unmasked. The interaction between induction and consolidation and maintenance was tested at a two-sided significance level of 0·05 by a stratified Cox regression model that included the interaction term between maintenance treatment and induction and consolidation treatment. Efficacy analyses were done in the maintenance-specific intention-to-treat population, which comprised all patients who underwent second randomisation. Safety was analysed in all patients in the daratumumab group who received at least one dose and all patients randomly assigned to observation only. This trial is registered with ClinicalTrials.gov, NCT02541383. Long-term follow-up is ongoing and the trial is closed to new participants., Findings: Between May 30, 2016, and June 18, 2018, 886 patients (458 [84%] of 543 in the D-VTd group and 428 [79%] of 542 in the VTd group) were randomly assigned to daratumumab maintenance (n=442) or observation only (n=444). At a median follow-up of 35·4 months (IQR 30·2-39·9) from second randomisation, median progression-free survival was not reached (95% CI not evaluable [NE]-NE) with daratumumab versus 46·7 months (40·0-NE) with observation only (hazard ratio 0·53, 95% CI 0·42-0·68, p<0·0001). A prespecified analysis of progression-free survival results showed a significant interaction between maintenance and induction and consolidation therapy (p<0·0001). The most common grade 3 or 4 adverse events were lymphopenia (16 [4%] of 440 patients in the daratumumab group vs eight [2%] of 444 patients in the observation-only group), hypertension (13 [3%] vs seven [2%]), and neutropenia (nine [2%] vs ten [2%]). Serious adverse events occurred in 100 (23%) patients in the daratumumab group and 84 (19%) patients in the observation-only group. In the daratumumab group, two adverse events led to death (septic shock and natural killer-cell lymphoblastic lymphoma); both were related to treatment., Interpretation: Daratumumab maintenance every 8 weeks for 2 years significantly reduced the risk of disease progression or death compared with observation only. Longer follow-up and other ongoing studies will shed further light on the optimal daratumumab-containing post-ASCT maintenance treatment strategy., Funding: Janssen Research & Development, the Intergroupe Francophone du Myélome, and the Dutch-Belgian Cooperative Trial Group for Hematology Oncology., Competing Interests: Declaration of interests PM reports personal fees from Celgene, Amgen, Takeda, Janssen, and AbbVie, outside the submitted work. CH reports personal fees from Janssen, AbbVie, Amgen, and Celgene, outside the submitted work. AP reports personal fees from Celgene, Amgen, Janssen, Sanofi, and Takeda, outside the submitted work. BA reports grants from Amgen, Celgene, Sanofi, and Janssen, during the conduct of the study; personal fees from Amgen, Celgene/Bristol Myers Squibb, Sanofi, GlaxoSmithKline, Takeda, and Janssen, outside the submitted work; and advisory board participation from Amgen, Celgene/Bristol Myers Squibb, Sanofi, GlaxoSmithKline, and Janssen, outside the submitted work. KB reports grants from Celgene outside the submitted work; personal fees from Celgene, Janssen, Takeda, and Amgen, outside the submitted work; and non-financial support from Celgene, AbbVie, and Takeda, outside the submitted work. SZ reports grants from Celgene, Janssen, and Takeda, during the conduct of the study. MD reports grants from Celgene and Janssen during the conduct of the study; participation on an advisory board for Celgene, Takeda, Janssen, Sanofi, and Oncopeptides, outside the submitted work. TD reports grants from Celgene and Janssen, during the conduct of the study; and personal fees and advisory board participation from Celgene, Takeda, Janssen, and Amgen, outside of the submitted work. CD reports personal fees from Janssen, outside the submitted work. TF reports personal fees from Janssen, Bristol Myers Squibb, Takeda, Amgen, Roche, Karyopharm, Sanofi, and Oncopeptides, outside the submitted work. CS reports personal fees from Celgene, outside the submitted work. MMo reports grants from Stemline, BMS, Amgen, Jazz Pharmaceuticals, Novartis, Takeda, Janssen, GSK, Sanofi, and Celgene; non-financial support from Stemline, BMS, Amgen, Jazz Pharmaceuticals, Novartis, Takeda, Janssen, GSK, Sanofi, and Celgene; and personal fees from Stemline, BMS, Amgen, Jazz Pharmaceuticals, Novartis, Takeda, Janssen, GSK, Sanofi, and Celgene, outside the submitted work. LK reports personal fees from Amgen, Janssen, Celgene, Takeda, and AbbVie, outside the submitted work. XL reports personal fees from Janssen, outside the submitted work. MMa reports personal fees from and advisory board participation for Amgen, Celgene, Janssen, and Takeda, outside the submitted work. A-MS reports personal fees from Celgene outside the submitted work. NWCJvdD reports grants from Amgen, Bristol Myers Squibb, Celgene, Janssen, and Novartis, during the conduct of the study; and personal fees from Amgen, Bristol Myers Squibb, Celgene, Janssen, Novartis, Bayer, Roche, Servier, and Takeda, outside the submitted work. AB reports personal fees from Amgen, Celgene, Janssen, and Bristol Myers Squibb, outside the submitted work. CT reports personal fees from Janssen, outside the submitter work. MR reports grants from Janssen, during the conduct of the study; personal fees and travel support from Celgene, Amgen, Sanofi, Takeda, and Janssen, outside the submitted work. M-DL reports grants from AbbVie, Amgen, Janssen, Roche, and Takeda, during the conduct of the study; and personal fees and advisory board participation from AbbVie, Janssen, Roche, and Takeda, outside the submitted work. TA reports employment and equity ownership from Genmab. MK reports employment with Janssen. KZ reports employment with Janssen. CdB reports employment and equity ownership from Janssen. SV reports employment with Janssen. TK reports employment with Janssen. VV reports employment with Janssen. JV reports employment with Janssen. HA-L reports grants from Celgene and Janssen, during the conduct of the study; and personal fees from Celgene, Amgen, Bristol-Myers Squibb, Sanofi, and Janssen, outside the submitted work. PS reports personal fees from Celgene and Janssen, outside the submitted work. All other authors declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
157. Peripheral neuropathy symptoms, pain, and functioning in previously treated multiple myeloma patients treated with selinexor, bortezomib, and dexamethasone.
- Author
-
Sanchez L, Leleu X, Beaumont JL, Yu H, Hudgens S, Simonova M, Auner HW, Quach H, Delimpasi S, Špička I, Pour L, Kriachok I, Dimopoulos MA, Usenko G, Hájek R, Benjamin R, Sinha DK, Venner C, Illmer T, Garg MK, Stevens DA, Jagannath S, Levy M, Anderson LD Jr, Bahlis NJ, Facon T, Cavo M, Chai Y, Ma X, Tang S, Leong H, Shah J, Shacham S, Kauffman M, Richardson P, and Grosicki S
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib therapeutic use, Dexamethasone therapeutic use, Female, Humans, Hydrazines therapeutic use, Male, Triazoles therapeutic use, Bortezomib adverse effects, Dexamethasone adverse effects, Hydrazines adverse effects, Multiple Myeloma drug therapy, Pain chemically induced, Peripheral Nervous System Diseases chemically induced, Triazoles adverse effects
- Published
- 2021
- Full Text
- View/download PDF
158. Quality of life analyses in patients with multiple myeloma: results from the Selinexor (KPT-330) Treatment of Refractory Myeloma (STORM) phase 2b study.
- Author
-
Tremblay G, Daniele P, Breeze J, Li L, Shah J, Shacham S, Kauffman M, Engelhardt M, Chari A, Nooka A, Vogl D, Gavriatopoulou M, Dimopoulos MA, Richardson P, Biran N, Siegel D, Vlummens P, Doyen C, Facon T, Mohty M, Meuleman N, Levy M, Costa L, Hoffman JE, Delforge M, Kaminetzky D, Weisel K, Raab M, Dingli D, Tuchman S, Laurent F, Vij R, Schiller G, Moreau P, Richter J, Schreder M, Podar K, Parker T, Cornell RF, Lionel K, Choquet S, and Sundar J
- Subjects
- Adult, Aged, Aged, 80 and over, Dexamethasone administration & dosage, Female, Follow-Up Studies, Humans, Hydrazines administration & dosage, Male, Middle Aged, Multiple Myeloma pathology, Neoplasm Recurrence, Local pathology, Prognosis, Survival Rate, Triazoles administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Drug Resistance, Neoplasm, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Quality of Life
- Abstract
Background: Selinexor is an oral, selective nuclear export inhibitor. STORM was a phase 2b, single-arm, open-label, multicenter trial of selinexor with low dose dexamethasone in patients with penta-exposed relapsed/refractory multiple myeloma (RRMM) that met its primary endpoint, with overall response of 26% (95% confidence interval [CI], 19 to 35%). Health-related quality of life (HRQoL) was a secondary endpoint measured using the Functional Assessment of Cancer Therapy - Multiple Myeloma (FACT-MM). This study examines impact of selinexor treatment on HRQoL of patients treated in STORM and reports two approaches to calculate minimal clinically important differences for the FACT-MM., Methods: FACT-MM data were collected at baseline, on day 1 of each 4-week treatment cycle, and at end of treatment (EOT). Changes from baseline were analyzed for the FACT-MM total score, FACT-trial outcome index (TOI), FACT-General (FACT-G), and the MM-specific domain using mixed-effects regression models. Two approaches for evaluating minimal clinically important differences were explored: the first defined as 10% of the instrument range, and the second based on estimated mean baseline differences between Eastern Cooperative Oncology Group performance status (ECOG PS) scores. Post-hoc difference analysis compared change in scores from baseline to EOT for treatment responders and non-responders., Results: Eighty patients were included in the analysis; the mean number of prior therapies was 7.9 (standard deviation [SD] 3.1), and mean duration of myeloma was 7.6 years (SD 3.4). Each exploratory minimal clinically important difference threshold yielded consistent results whereby most patients did not experience HRQoL decline during the first six cycles of treatment (range: 53.9 to 75.7% for the first approach; range: 52.6 to 72.9% for the second). Treatment responders experienced less decline in HRQoL from baseline to EOT than non-responders, which was significant for the FACT-G, but not for other scores., Conclusion: The majority of patients did not experience decline in HRQoL based on minimal clinically important differences during early cycles of treatment with selinexor and dexamethasone in the STORM trial. An anchor-based approach utilizing patient-level data (ECOG PS score) to define minimal clinically important differences for the FACT-MM gave consistent results with a distribution-based approach., Trial Registration: This trial was registered on ClinicalTrials.gov under the trial-ID NCT02336815 on January 8, 2015., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
159. Selinexor, bortezomib, and dexamethasone versus bortezomib and dexamethasone in previously treated multiple myeloma: Outcomes by cytogenetic risk.
- Author
-
Richard S, Chari A, Delimpasi S, Simonova M, Spicka I, Pour L, Kriachok I, Dimopoulos MA, Pylypenko H, Auner HW, Leleu X, Usenko G, Hajek R, Benjamin R, Dolai TK, Sinha DK, Venner CP, Garg M, Stevens DA, Quach H, Jagannath S, Moreau P, Levy M, Badros A, Anderson LD Jr, Bahlis NJ, Facon T, Mateos MV, Cavo M, Chang H, Landesman Y, Chai Y, Arazy M, Shah J, Shacham S, Kauffman MG, Grosicki S, and Richardson PG
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib adverse effects, Cytogenetic Analysis, Dexamethasone adverse effects, Female, Humans, Hydrazines adverse effects, Male, Middle Aged, Multiple Myeloma genetics, Progression-Free Survival, Treatment Outcome, Triazoles adverse effects, Young Adult, Antineoplastic Agents therapeutic use, Bortezomib therapeutic use, Dexamethasone therapeutic use, Hydrazines therapeutic use, Multiple Myeloma drug therapy, Triazoles therapeutic use
- Abstract
In the phase 3 BOSTON study, patients with multiple myeloma (MM) after 1-3 prior regimens were randomized to once-weekly selinexor (an oral inhibitor of exportin 1 [XPO1]) plus bortezomib-dexamethasone (XVd) or twice-weekly bortezomib-dexamethasone (Vd). Compared with Vd, XVd was associated with significant improvements in median progression-free survival (PFS), overall response rate (ORR), and lower rates of peripheral neuropathy, with trends in overall survival (OS) favoring XVd. In BOSTON, 141 (35.1%) patients had MM with high-risk (presence of del[17p], t[4;14], t[14;16], or ≥4 copies of amp1q21) cytogenetics (XVd, n = 70; Vd, n = 71), and 261 (64.9%) exhibited standard-risk cytogenetics (XVd, n = 125; Vd, n = 136). Among patients with high-risk MM, median PFS was 12.91 months for XVd and 8.61 months for Vd (HR, 0.73 [95% CI, (0.4673, 1.1406)], p = 0.082), and ORRs were 78.6% and 57.7%, respectively (OR 2.68; p = 0.004). In the standard-risk subgroup, median PFS was 16.62 months for XVd and 9.46 months for Vd (HR 0.61; p = 0.004), and ORRs were 75.2% and 64.7%, respectively (OR 1.65; p = 0.033). The safety profiles of XVd and Vd in both subgroups were consistent with the overall population. These data suggest that selinexor can confer benefits to patients with MM regardless of cytogenetic risk. ClinicalTrials.gov identifier: NCT03110562., (© 2021 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
160. Effective anti-BCMA retreatment in multiple myeloma.
- Author
-
Gazeau N, Beauvais D, Yakoub-Agha I, Mitra S, Campbell TB, Facon T, and Manier S
- Subjects
- B-Cell Maturation Antigen, Humans, Retreatment, T-Lymphocytes, Tumor Microenvironment, Antibodies, Bispecific therapeutic use, Multiple Myeloma drug therapy
- Abstract
The recent emergence of anti-B-cell maturation antigen (BCMA) therapies holds great promise in multiple myeloma (MM). These include chimeric antigen receptor (CAR) T cells, bispecific antibodies, and antibody-drug conjugates. Their development in clinical trials and further approval are changing the strategy for treating MM. Considering that a cure has not been reached, a central question in the coming years will be the possibility of using these therapies sequentially. Here, we report 2 cases of the serial use of anti-BCMA therapies with parallel monitoring of BCMA expression and anti-CAR antibodies. We further discuss recent data from clinical studies that have informed us about the different mechanisms of resistance to anti-BCMA therapies, including antigen escape, BCMA shedding, anti-drug antibodies, T-cell exhaustion, and the emergence of an immunosuppressive microenvironment. This knowledge will be essential to help guide the strategy of serial treatments with anti-BCMA therapies., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
161. The DNA methylation landscape of multiple myeloma shows extensive inter- and intrapatient heterogeneity that fuels transcriptomic variability.
- Author
-
Derrien J, Guérin-Charbonnel C, Gaborit V, Campion L, Devic M, Douillard E, Roi N, Avet-Loiseau H, Decaux O, Facon T, Mallm JP, Eils R, Munshi NC, Moreau P, Herrmann C, Magrangeas F, and Minvielle S
- Subjects
- Computational Biology methods, CpG Islands, Disease Susceptibility, Epigenomics methods, Gene Expression Profiling, Histones metabolism, Humans, Molecular Sequence Annotation, Multiple Myeloma diagnosis, Multiple Myeloma metabolism, Multiple Myeloma mortality, Prognosis, Promoter Regions, Genetic, DNA Methylation, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Genetic Heterogeneity, Multiple Myeloma genetics, Transcriptome
- Abstract
Background: Cancer evolution depends on epigenetic and genetic diversity. Historically, in multiple myeloma (MM), subclonal diversity and tumor evolution have been investigated mostly from a genetic perspective., Methods: Here, we performed an analysis of 42 MM samples from 21 patients by using enhanced reduced representation bisulfite sequencing (eRRBS). We combined several metrics of epigenetic heterogeneity to analyze DNA methylation heterogeneity in MM patients., Results: We show that MM is characterized by the continuous accumulation of stochastic methylation at the promoters of development-related genes. High combinatorial entropy change is associated with poor outcomes in our pilot study and depends predominantly on partially methylated domains (PMDs). These PMDs, which represent the major source of inter- and intrapatient DNA methylation heterogeneity in MM, are linked to other key epigenetic aberrations, such as CpG island (CGI)/transcription start site (TSS) hypermethylation and H3K27me3 redistribution as well as 3D organization alterations. In addition, transcriptome analysis revealed that intratumor methylation heterogeneity was associated with low-level expression and high variability., Conclusions: We propose that disrupted DNA methylation in MM is responsible for high epigenetic and transcriptomic instability allowing tumor cells to adapt to environmental changes by tapping into a pool of evolutionary trajectories., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
162. Carfilzomib, dexamethasone and daratumumab in relapsed or refractory multiple myeloma: results of the phase III study CANDOR by prior lines of therapy.
- Author
-
Quach H, Nooka A, Samoylova O, Venner CP, Kim K, Facon T, Spencer A, Usmani SZ, Grosicki S, Suzuki K, Delimpasi S, Weisel K, Obreja M, Zahlten-Kumeli A, and Mateos MV
- Subjects
- Humans, Neoplasm Recurrence, Local drug therapy, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Oligopeptides therapeutic use
- Published
- 2021
- Full Text
- View/download PDF
163. Stem cell yield and transplantation in transplant-eligible newly diagnosed multiple myeloma patients receiving daratumumab + bortezomib/thalidomide/dexamethasone in the phase 3 CASSIOPEIA study.
- Author
-
Hulin C, Offner F, Moreau P, Roussel M, Belhadj K, Benboubker L, Caillot D, Facon T, Garderet L, Kuhnowski F, Stoppa AM, Kolb B, Tiab M, Jie KS, Westerman M, Lambert J, Pei L, Vanquickelberghe V, De Boer C, Vermeulen J, Kampfenkel T, Sonneveld P, and Van de Donk NWCJ
- Subjects
- Antibodies, Monoclonal, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib therapeutic use, Dexamethasone therapeutic use, Humans, Stem Cell Transplantation, Stem Cells, Transplantation, Autologous, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy, Thalidomide therapeutic use
- Published
- 2021
- Full Text
- View/download PDF
164. Up-front carfilzomib, lenalidomide, and dexamethasone with transplant for patients with multiple myeloma: the IFM KRd final results.
- Author
-
Roussel M, Lauwers-Cances V, Wuilleme S, Belhadj K, Manier S, Garderet L, Escoffre-Barbe M, Mariette C, Benboubker L, Caillot D, Sonntag C, Touzeau C, Dupuis J, Moreau P, Leleu X, Facon T, Hébraud B, Corre J, and Attal M
- Subjects
- Combined Modality Therapy, Dexamethasone adverse effects, Disease-Free Survival, Female, Humans, Kaplan-Meier Estimate, Lenalidomide adverse effects, Male, Middle Aged, Neoplasm, Residual pathology, Oligopeptides adverse effects, Survival Analysis, Treatment Outcome, Dexamethasone therapeutic use, Hematopoietic Stem Cell Transplantation, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Oligopeptides therapeutic use
- Abstract
Bortezomib, lenalidomide, and dexamethasone plus transplant is a standard of care for eligible patients with multiple myeloma. Because responses can deepen with time, regimens with longer and more potent induction/consolidation phases are needed. In this phase 2 study, patients received eight 28-day cycles of carfilzomib (K) 20/36 mg/m2 (days 1-2, 8-9, 15-16), lenalidomide (R) 25 mg (days 1-21), and dexamethasone (d) 20 mg (days 1-2, 8-9, 15-16, 22-23). All patients proceeded to transplant after 4 cycles and received 1 year of lenalidomide maintenance (10 mg, days 1-21). The primary objective was stringent complete response at the completion of consolidation. Overall, 48 patients were screened and 46 enrolled; 21% had adverse cytogenetics. Among 42 evaluable patients after consolidation, 26 were in stringent complete response (CR; 61.9%), 27 were at least in CR (64.3%): 92.6% had undetectable minimal residual disease according to flow cytometry (≥2.5 × 10-5) and 63.0% according to next-generation sequencing (10-6). Median time to CR was 10.6 months. According to multiparametric flow cytometry and next-generation sequencing, 69.0% and 66.7% of patients, respectively, had undetectable minimal residual disease at some point. With a median follow-up of 60.5 months, 21 patients progressed, and 10 died (7 of multiple myeloma). Median progression-free survival was 56.4 months. There were no KRd-related deaths. Four patients discontinued the program due to toxicities; 56 serious adverse events were reported in 31 patients, including 8 cardiovascular events (2 heart failures, 5 pulmonary embolisms or deep vein thrombosis). Common grade 3/4 adverse events were hematologic (74%) and infectious (22%). In summary, 8 cycles of KRd produce fast and deep responses in transplant-eligible patients with newly diagnosed multiple myeloma. The safety profile is acceptable, but cardiovascular adverse events should be closely monitored. This clinical trial is registered at www.clinicaltrials.gov as #NCT02405364., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
165. Oral ixazomib, lenalidomide, and dexamethasone for transplant-ineligible patients with newly diagnosed multiple myeloma.
- Author
-
Facon T, Venner CP, Bahlis NJ, Offner F, White DJ, Karlin L, Benboubker L, Rigaudeau S, Rodon P, Voog E, Yoon SS, Suzuki K, Shibayama H, Zhang X, Twumasi-Ankrah P, Yung G, Rifkin RM, Moreau P, Lonial S, Kumar SK, Richardson PG, and Rajkumar SV
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Boron Compounds administration & dosage, Boron Compounds adverse effects, Dexamethasone administration & dosage, Dexamethasone adverse effects, Disease-Free Survival, Double-Blind Method, Female, Follow-Up Studies, Glycine administration & dosage, Glycine adverse effects, Glycine analogs & derivatives, Humans, Lenalidomide administration & dosage, Lenalidomide adverse effects, Male, Middle Aged, Survival Rate, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy, Multiple Myeloma mortality
- Abstract
Continuous lenalidomide-dexamethasone (Rd)-based regimens are among the standards of care in transplant-ineligible newly diagnosed multiple myeloma (NDMM) patients. The oral proteasome inhibitor ixazomib is suitable for continuous dosing, with predictable, manageable toxicities. In the double-blind, placebo-controlled TOURMALINE-MM2 trial, transplant-ineligible NDMM patients were randomized to ixazomib 4 mg (n = 351) or placebo (n = 354) plus Rd. After 18 cycles, dexamethasone was discontinued and treatment was continued using reduced-dose ixazomib (3 mg) and lenalidomide (10 mg) until progression/toxicity. The primary endpoint was progression-free survival (PFS). Median PFS was 35.3 vs 21.8 months with ixazomib-Rd vs placebo-Rd, respectively (hazard ratio [HR], 0.830; 95% confidence interval, 0.676-1.018; P = .073; median follow-up, 53.3 and 55.8 months). Complete (26% vs 14%; odds ratio [OR], 2.10; P < .001) and ≥ very good partial response (63% vs 48%; OR, 1.87; P < .001) rates were higher with ixazomib-Rd vs placebo-Rd. In a prespecified high-risk cytogenetics subgroup, median PFS was 23.8 vs 18.0 months (HR, 0.690; P = .019). Overall, treatment-emergent adverse events (TEAEs) were mostly grade 1/2. With ixazomib-Rd vs placebo-Rd, 88% vs 81% of patients experienced grade ≥3 TEAEs, 66% vs 62% serious TEAEs, and 35% vs 27% TEAEs resulting in regimen discontinuation; 8% vs 6% died on study. Addition of ixazomib to Rd was tolerable with no new safety signals and led to a clinically meaningful PFS benefit of 13.5 months. Ixazomib-Rd is a feasible option for certain patients who can benefit from an all-oral triplet combination. This trial was registered at www.clinicaltrials.gov as #NCT01850524., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
166. Subgroup analysis of ICARIA-MM study in relapsed/refractory multiple myeloma patients with high-risk cytogenetics.
- Author
-
Harrison SJ, Perrot A, Alegre A, Simpson D, Wang MC, Spencer A, Delimpasi S, Hulin C, Sunami K, Facon T, Vlummens P, Yong K, Campana F, Inchauspé M, Macé S, Risse ML, van de Velde H, and Richardson P
- Subjects
- Abnormal Karyotype, Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Chromosomes, Human, Pair 1 genetics, Dexamethasone administration & dosage, Febrile Neutropenia chemically induced, Female, Humans, Immunologic Factors administration & dosage, Kaplan-Meier Estimate, Male, Middle Aged, Multiple Myeloma genetics, Multiple Myeloma mortality, Myeloma Proteins analysis, Pneumonia chemically induced, Recurrence, Risk, Thalidomide administration & dosage, Thalidomide analogs & derivatives, Trisomy, Clinical Trials, Phase III as Topic statistics & numerical data, Multiple Myeloma drug therapy, Randomized Controlled Trials as Topic statistics & numerical data, Salvage Therapy
- Abstract
Treatment benefit in multiple myeloma (MM) patients with high-risk cytogenetics remains suboptimal. The phase 3 ICARIA-MM trial (NCT02990338) showed that isatuximab plus pomalidomide-dexamethasone prolongs median progression-free survival (mPFS) in patients with relapsed/refractory MM (RRMM). This subgroup analysis of ICARIA-MM compared the benefit of isatuximab in high-risk [defined by the presence of del(17p), t(4;14) or t(14;16)] versus standard-risk patients. The efficacy of isatuximab in patients with gain(1q21) abnormality was also assessed in a retrospective subgroup analysis. In ICARIA-MM, 307 patients received isatuximab-pomalidomide-dexamethasone (n = 154) or pomalidomide-dexamethasone (n = 153). Isatuximab (10 mg/kg intravenously) was given weekly in the first 28-day cycle, and every other week thereafter. Standard pomalidomide-dexamethasone doses were given. Isatuximab-pomalidomide-dexamethasone improved mPFS (7·5 vs 3·7 months; HR, 0·66; 95% CI, 0·33-1·28) and overall response rate (ORR, 50·0% vs 16·7%) in high-risk patients. In patients with isolated gain(1q21), isatuximab addition improved mPFS (11·2 vs 4·6 months; HR, 0·50; 95% CI, 0·28-0·88) and ORR (53·6% vs 27·6%). More grade ≥3 adverse events occurred in high-risk patients receiving isatuximab (95·7%) versus the control group (67·6%); however, isatuximab did not increase events leading to discontinuation or treatment-related mortality. Isatuximab-pomalidomide-dexamethasone provides a consistent benefit over pomalidomide-dexamethasone treatment in RRMM patients regardless of cytogenetic risk., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
167. Isatuximab, carfilzomib, and dexamethasone in relapsed multiple myeloma (IKEMA): a multicentre, open-label, randomised phase 3 trial.
- Author
-
Moreau P, Dimopoulos MA, Mikhael J, Yong K, Capra M, Facon T, Hajek R, Špička I, Baker R, Kim K, Martinez G, Min CK, Pour L, Leleu X, Oriol A, Koh Y, Suzuki K, Risse ML, Asset G, Macé S, and Martin T
- Subjects
- Administration, Intravenous, Aged, Drug Therapy, Combination, Female, Humans, Male, Middle Aged, Progression-Free Survival, Prospective Studies, Recurrence, Thalidomide therapeutic use, Anti-Inflammatory Agents therapeutic use, Antibodies, Monoclonal, Humanized therapeutic use, Dexamethasone therapeutic use, Immunologic Factors therapeutic use, Multiple Myeloma drug therapy, Oligopeptides therapeutic use, Thalidomide analogs & derivatives
- Abstract
Background: Isatuximab is an anti-CD38 monoclonal antibody approved in combination with pomalidomide-dexamethasone and carfilzomib-dexamethasone for relapsed or refractory multiple myeloma. This phase 3, open-label study compared the efficacy of isatuximab plus carfilzomib-dexamethasone versus carfilzomib-dexamethasone in patients with relapsed multiple myeloma., Methods: This was a prospective, randomised, open-label, parallel-group, phase 3 study done at 69 study centres in 16 countries across North America, South America, Europe, and the Asia-Pacific region. Patients with relapsed or refractory multiple myeloma aged at least 18 years who had received one to three previous lines of therapy and had measurable serum or urine M-protein were eligible. Patients were randomly assigned (3:2) to isatuximab plus carfilzomib-dexamethasone (isatuximab group) or carfilzomib-dexamethasone (control group). Patients in the isatuximab group received isatuximab 10 mg/kg intravenously weekly for the first 4 weeks, then every 2 weeks. Both groups received the approved schedule of intravenous carfilzomib and oral or intravenous dexamethasone. Treatment continued until progression or unacceptable toxicity. The primary endpoint was progression-free survival and was assessed in the intention-to-treat population according to assigned treatment. Safety was assessed in all patients who received at least one dose according to treatment received. The study is registered at ClinicalTrials.gov, NCT03275285., Findings: Between Nov 15, 2017, and March 21, 2019, 302 patients with a median of two previous lines of therapy were enrolled. 179 were randomly assigned to the isatuximab group and 123 to the control group. Median progression-free survival was not reached in the isatuximab group compared with 19·15 months (95% CI 15·77-not reached) in the control group, with a hazard ratio of 0·53 (99% CI 0·32-0·89; one-sided p=0·0007). Treatment-emergent adverse events (TEAEs) of grade 3 or worse occurred in 136 (77%) of 177 patients in the isatuximab group versus 82 (67%) of 122 in the control group, serious TEAEs occurred in 105 (59%) versus 70 (57%) patients, and TEAEs led to discontinuation in 15 (8%) versus 17 (14%) patients. Fatal TEAEs during study treatment occurred in six (3%) versus four (3%) patients., Interpretation: The addition of isatuximab to carfilzomib-dexamethasone significantly improves progression-free survival and depth of response in patients with relapsed multiple myeloma, representing a new standard of care for this patient population., Funding: Sanofi. VIDEO ABSTRACT., Competing Interests: Declaration of interests PM reports honoraria from Amgen, Celgene, Janssen, Novartis, and Takeda; and a consulting or advisory role for Amgen, Celgene, Janssen, Novartis, and Takeda. M-AD reports consulting or advisory role for Amgen, Bristol Myers Squibb (BMS), Celgene, Janssen, and Takeda. JM reports honoraria from Celgene, Takeda, BMS, Janssen, and Amgen; and a consulting or advisory role for Amgen, BMS, Celgene, Janssen-Cilag, and Takeda. KY reports a consulting or advisory role for Amgen, Janssen, and Takeda; speaker's bureau for Amgen and Takeda; and research funding from Amgen and Sanofi. MC reports speaker's bureau for Amgen, Janssen, and Sanofi. TF reports an advisory role for Amgen, BMS, Celgene, Karyopharm, Oncopeptides, Roche, Sanofi, and Takeda; and speaker's bureau for Takeda. RH reports personal fees from AbbVie, Amgen, BMS, Celgene, Pharma Mar, Novartis, and Takeda; and grants from Novartis and Takeda. IS reports a consulting or advisory role for Amgen, BMS, Celgene, Janssen-Cilag, Novartis, and Takeda; and speakers' bureau for Amgen, BMS, Celgene, Janssen-Cilag, Novartis, and Takeda. TM reports research funding from Amgen and Sanofi. M-LR, GA, and SM are employed by Sanofi and may hold shares or stock options in the company. RB reports research funding from AbbVie, Acerta Pharma, Alexion, Amgen, Bayer, BMS, Boehringer Ingelheim, Celgene, CSL Behring, Daiichi Sankyo, Jansen-Cilag, MorphoSys, Pfizer, Portola, Rigel Pharmaceuticals, Roche, Sanofi, Takeda, and Technoclone; and consulting or advisory roles for Jansen-Cilag and Roche. KK reports research funding from BMS and Janssen; and honoraria from Amgen, BMS, Janssen, and Takeda. AO reports honoraria from Amgen, Celgene, and Janssen. KS reports honoraria from AbbVie, Amgen, BMS, Celgene, Jansen, Novartis, ONO, Sanofi, and Takeda; and consulting or advisory roles for AbbVie, BMS, and Celgene. All other authors declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
168. Effect of age and frailty on the efficacy and tolerability of once-weekly selinexor, bortezomib, and dexamethasone in previously treated multiple myeloma.
- Author
-
Auner HW, Gavriatopoulou M, Delimpasi S, Simonova M, Spicka I, Pour L, Dimopoulos MA, Kriachok I, Pylypenko H, Leleu X, Doronin V, Usenko G, Hajek R, Benjamin R, Dolai TK, Sinha DK, Venner CP, Garg M, Stevens DA, Quach H, Jagannath S, Moreau P, Levy M, Badros A, Anderson LD Jr, Bahlis NJ, Facon T, Mateos MV, Cavo M, Chai Y, Arazy M, Shah J, Shacham S, Kauffman MG, Richardson PG, and Grosicki S
- Subjects
- Adult, Age Factors, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib administration & dosage, Clinical Trials, Phase III as Topic statistics & numerical data, Dexamethasone administration & dosage, Drug Administration Schedule, Female, Frailty diagnosis, Gastrointestinal Diseases chemically induced, Hematologic Diseases chemically induced, Humans, Hydrazines administration & dosage, Kaplan-Meier Estimate, Male, Middle Aged, Multicenter Studies as Topic statistics & numerical data, Multiple Myeloma complications, Peripheral Nervous System Diseases chemically induced, Progression-Free Survival, Randomized Controlled Trials as Topic statistics & numerical data, Retrospective Studies, Severity of Illness Index, Triazoles administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib adverse effects, Dexamethasone adverse effects, Frailty complications, Hydrazines adverse effects, Multiple Myeloma drug therapy, Triazoles adverse effects
- Abstract
Elderly and frail patients with multiple myeloma (MM) are more vulnerable to the toxicity of combination therapies, often resulting in treatment modifications and suboptimal outcomes. The phase 3 BOSTON study showed that once-weekly selinexor and bortezomib with low-dose dexamethasone (XVd) improved PFS and ORR compared with standard twice-weekly bortezomib and moderate-dose dexamethasone (Vd) in patients with previously treated MM. This is a retrospective subgroup analysis of the multicenter, prospective, randomized BOSTON trial. Post hoc analyses were performed to compare XVd versus Vd safety and efficacy according to age and frailty status (<65 and ≥65 years, nonfrail and frail). Patients ≥65 years with XVd had higher ORR (OR 1.77, p = .024), ≥VGPR (OR, 1.68, p = .027), PFS (HR 0.55, p = .002), and improved OS (HR 0.63, p = .030), compared with Vd. In frail patients, XVd was associated with a trend towards better PFS (HR 0.69, p = .08) and OS (HR 0.62, p = .062). Significant improvements were also observed in patients <65 (ORR and TTNT) and nonfrail patients (PFS, ORR, ≥VGPR, and TTNT). Patients treated with XVd had a lower incidence of grade ≥ 2 peripheral neuropathy in ≥65 year-old (22% vs. 37%; p = .0060) and frail patients (15% vs. 44%; p = .0002). Grade ≥3 TEAEs were not observed more often in older compared to younger patients, nor in frail compared to nonfrail patients. XVd is safe and effective in patients <65 and ≥65 and in nonfrail and frail patients with previously treated MM., (© 2021 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
169. Case Report: Two Cases of Cryptosporidiosis in Heavily Pretreated Patients With Myeloma.
- Author
-
Demonchy J, Cordier C, Fréalle E, Demarquette H, Herbaux C, Escure G, Willaume A, Van De Wyngaert Z, Noel MP, Facon T, Faure K, Caro J, Morgan G, Davies FE, Alfandari S, Bories C, and Boyle EM
- Subjects
- Antiparasitic Agents therapeutic use, Combined Modality Therapy adverse effects, Combined Modality Therapy methods, Cryptosporidiosis therapy, Cryptosporidium, Diarrhea diagnosis, Diarrhea etiology, Diarrhea therapy, Disease Management, Disease Susceptibility, Female, Humans, Male, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma therapy, Nitro Compounds therapeutic use, Retreatment, Symptom Assessment, Thiazoles therapeutic use, Tomography, X-Ray Computed, Treatment Outcome, Cryptosporidiosis diagnosis, Cryptosporidiosis etiology, Multiple Myeloma complications
- Published
- 2021
- Full Text
- View/download PDF
170. Association of Morbid Progression With Overall Survival Among Patients With Multiple Myeloma: Validation of the Progression-free Survival Endpoint.
- Author
-
Rosenberg AS, Facon T, Parikh K, Chung W, Srinivasan S, Kotey S, and Tuscano J
- Subjects
- Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Multiple Myeloma physiopathology, Progression-Free Survival, Hypercalcemia etiology, Multiple Myeloma mortality
- Abstract
Introduction: Multiple myeloma (MM) is an incurable malignancy, marked by end-organ damage that is frequently irreversible. Progressive disease (PD) can be defined as morbid PD, associated with new-onset hypercalcemia, renal insufficiency, anemia, or lytic bone lesions (CRAB symptoms), or as asymptomatic biochemical progression. The frequency of morbid versus asymptomatic PD and its effect on survival is unknown. Our aim was to determine the incidence of morbid PD, and to evaluate if this influences survival., Patients and Methods: Data from 2 phase III trials of transplant-ineligible patients with newly diagnosed MM were included in a post hoc analysis., Results: Of 2082 patients enrolled, 1243 (59.7%) experienced PD. At first progression, 543 (43.7%) patients had morbid PD; 12 (2.2%) had hypercalcemia, 271 (49.9%) had renal insufficiency, 370 (68.1%) developed anemia, and 79 (14.5%) developed new or enlarged bone lesions. A total of 700 (56.3%) patients had asymptomatic PD. Patients with morbid PD had worse second progression-free survival (PFS) versus patients with asymptomatic biochemical PD (median second PFS, 11.5 months vs. 20.0 months; hazard ratio, 1.63; 95% confidence interval, 1.43-1.85; P < .0001) and worse overall survival (OS) (median OS, 23.2 months vs 39.3 months; hazard ratio, 1.51; 95% confidence interval, 1.30, 1.74; P < .0001)., Conclusions: Morbid PD occurs frequently and is associated with inferior second PFS and OS. As CRAB symptoms may not reverse with therapy, morbid PD is a meaningful event, and its association with a shortened PFS adds validity to PFS as a relevant endpoint in patients with MM., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
171. Can palliative care consultation increase integration of palliative care for patients with hematologic malignancies?
- Author
-
Prod'homme C, Touzet L, Pierrat M, Chevalier L, Lesaffre H, Berthon C, Coiteux V, Barbieux S, Beauvais D, Bauschert L, De Charette M, Goursaud L, Manier S, and Facon T
- Subjects
- Humans, Referral and Consultation, Hematologic Neoplasms therapy, Palliative Care
- Published
- 2021
- Full Text
- View/download PDF
172. Effect of prior treatments on selinexor, bortezomib, and dexamethasone in previously treated multiple myeloma.
- Author
-
Mateos MV, Gavriatopoulou M, Facon T, Auner HW, Leleu X, Hájek R, Dimopoulos MA, Delimpasi S, Simonova M, Špička I, Pour L, Kriachok I, Pylypenko H, Doronin V, Usenko G, Benjamin R, Dolai TK, Sinha DK, Venner CP, Garg M, Stevens DA, Quach H, Jagannath S, Moreau P, Levy M, Badros AZ, Anderson LD Jr, Bahlis NJ, Cavo M, Chai Y, Jeha J, Arazy M, Shah J, Shacham S, Kauffman MG, Richardson PG, and Grosicki S
- Subjects
- Antineoplastic Combined Chemotherapy Protocols pharmacology, Bortezomib pharmacology, Dexamethasone pharmacology, Female, Humans, Hydrazines pharmacology, Male, Multiple Myeloma pathology, Triazoles pharmacology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bortezomib therapeutic use, Dexamethasone therapeutic use, Hydrazines therapeutic use, Multiple Myeloma drug therapy, Triazoles therapeutic use
- Abstract
Therapeutic regimens for previously treated multiple myeloma (MM) may not provide prolonged disease control and are often complicated by significant adverse events, including peripheral neuropathy. In patients with previously treated MM in the Phase 3 BOSTON study, once weekly selinexor, once weekly bortezomib, and 40 mg dexamethasone (XVd) demonstrated a significantly longer median progression-free survival (PFS), higher response rates, deeper responses, a trend to improved survival, and reduced incidence and severity of bortezomib-induced peripheral neuropathy when compared with standard twice weekly bortezomib and 80 mg dexamethasone (Vd). The pre-specified analyses described here evaluated the influence of the number of prior lines of therapy, prior treatment with lenalidomide, prior proteasome inhibitor (PI) therapy, prior immunomodulatory drug therapy, and prior autologous stem cell transplant (ASCT) on the efficacy and safety of XVd compared with Vd. In this 1:1 randomized study, enrolled patients were assigned to receive once weekly oral selinexor (100 mg) with once weekly subcutaneous bortezomib (1.3 mg/m
2 ) and 40 mg per week dexamethasone (XVd) versus standard twice weekly bortezomib and 80 mg per week dexamethasone (Vd). XVd significantly improved PFS, overall response rate, time-to-next-treatment, and showed reduced all grade and grade ≥ 2 peripheral neuropathy compared with Vd regardless of prior treatments, but the benefits of XVd over Vd were more pronounced in patients treated earlier in their disease course who had either received only one prior therapy, had never been treated with a PI, or had prior ASCT. Treatment with XVd improved outcomes as compared to Vd regardless of prior therapies as well as manageable and generally reversible adverse events. XVd was associated with clinical benefit and reduced peripheral neuropathy compared to standard Vd in previously treated MM. These results suggest that the once weekly XVd regimen may be optimally administered to patients earlier in their course of disease, as their first bortezomib-containing regimen, and in those relapsing after ASCT.Trial registration: ClinicalTrials.gov (NCT03110562). Registered 12 April 2017. https://clinicaltrials.gov/ct2/show/NCT03110562 .- Published
- 2021
- Full Text
- View/download PDF
173. Targeting BCL-2 with venetoclax and dexamethasone in patients with relapsed/refractory t(11;14) multiple myeloma.
- Author
-
Kaufman JL, Gasparetto C, Schjesvold FH, Moreau P, Touzeau C, Facon T, Boise LH, Jiang Y, Yang X, Dunbar F, Vishwamitra D, Unger S, Macartney T, Pesko J, Yu Y, Salem AH, Ross JA, Hong WJ, Maciag PC, Pauff JM, and Kumar S
- Subjects
- Aged, Antibodies, Monoclonal pharmacology, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Bone Marrow pathology, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Bridged Bicyclo Compounds, Heterocyclic pharmacokinetics, Chromosomes, Human, Pair 11 genetics, Chromosomes, Human, Pair 14 genetics, Combined Modality Therapy, Dexamethasone administration & dosage, Female, Follow-Up Studies, Genes, bcl-2, Hematologic Diseases chemically induced, Hematopoietic Stem Cell Transplantation, Humans, Infections etiology, Kaplan-Meier Estimate, Male, Middle Aged, Multiple Myeloma genetics, Multiple Myeloma therapy, Recurrence, Signal Transduction, Sulfonamides administration & dosage, Sulfonamides adverse effects, Sulfonamides pharmacokinetics, Translocation, Genetic, bcl-X Protein, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Multiple Myeloma drug therapy, Neoplasm Proteins antagonists & inhibitors, Proto-Oncogene Proteins c-bcl-2 antagonists & inhibitors, Salvage Therapy, Sulfonamides pharmacology
- Abstract
Venetoclax (Ven) is a selective small-molecule inhibitor of BCL-2 that exhibits antitumoral activity against MM cells with t(11;14) translocation. We evaluated the safety and efficacy of Ven and dexamethasone (VenDex) combination in patients with t(11;14) positive relapsed/refractory (R/R) multiple myeloma (MM). This open-label, multicenter study had two distinct phases (phase one [P1], phase two [P2]). Patients in both phases received VenDex (oral Ven 800 mg/day + oral Dex 40 mg [20 mg for patients ≥75 years] on days 1, 8, and 15, per 21-day cycle). The primary objective of the P1 VenDex cohort was to assess safety and pharmacokinetics. Phase two further evaluated efficacy with objective response rate (ORR) and very good partial response or better. Correlative studies explored baseline BCL2 (BCL-2) and BCL2L1 (BCL-X
L ) gene expression, cytogenetics, and recurrent somatic mutations in MM. Twenty and 31 patients in P1 and P2 with t(11;14) positive translocation received VenDex. P1/P2 patients had received a median of 3/5 lines of prior therapy, and 20%/87% were refractory to daratumumab. Predominant grade 3/4 hematological adverse events (AEs) with ≥10% occurrence included lymphopenia (20%/19%), neutropenia (15%/7%), thrombocytopenia (10%/10%), and anemia (5%/16%). At a median follow-up of 12.3/9.2 months, ORR was 60%/48%. The duration of response estimate at 12 months was 50%/61%, and the median time to progression was 12.4/10.8 months. In biomarker evaluable patients, response to VenDex was independent of concurrent del(17p) or gain(1q) and mutations in key oncogenic signaling pathways, including MAPK and NF-kB. VenDex demonstrated efficacy and manageable safety in heavily-pre-treated patients with t(11;14) R/R MM., (© 2020 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)- Published
- 2021
- Full Text
- View/download PDF
174. Isatuximab plus pomalidomide and dexamethasone in elderly patients with relapsed/refractory multiple myeloma: ICARIA-MM subgroup analysis.
- Author
-
Schjesvold FH, Richardson PG, Facon T, Alegre A, Spencer A, Jurczyszyn A, Sunami K, Frenzel L, Min CK, Guillonneau S, Lin PL, Le-Guennec S, Campana F, van de Velde H, Bensfia S, and Bringhen S
- Subjects
- Aged, Antibodies, Monoclonal, Humanized, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Humans, Neoplasm Recurrence, Local drug therapy, Thalidomide analogs & derivatives, Thalidomide therapeutic use, Multiple Myeloma drug therapy
- Published
- 2021
- Full Text
- View/download PDF
175. Predictive biomarkers with isatuximab plus pomalidomide and dexamethasone in relapsed/refractory multiple myeloma.
- Author
-
Richardson PG, Facon T, Bensinger WI, Leleu X, Campana F, Macé S, Chiron M, van de Velde H, and Mikhael J
- Subjects
- Biomarkers, Tumor analysis, Humans, Multiple Myeloma diagnosis, Neoplasm Recurrence, Local diagnosis, Prognosis, Thalidomide therapeutic use, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Thalidomide analogs & derivatives
- Published
- 2021
- Full Text
- View/download PDF
176. del(17p) without TP53 mutation confers a poor prognosis in intensively treated newly diagnosed patients with multiple myeloma.
- Author
-
Corre J, Perrot A, Caillot D, Belhadj K, Hulin C, Leleu X, Mohty M, Facon T, Buisson L, Do Souto L, Lannes R, Dufrechou S, Prade N, Orsini-Piocelle F, Voillat L, Jaccard A, Karlin L, Macro M, Brechignac S, Dib M, Sanhes L, Fontan J, Clement-Filliatre L, Marolleau JP, Minvielle S, Moreau P, and Avet-Loiseau H
- Subjects
- Adult, Aged, Female, Humans, Male, Middle Aged, Multiple Myeloma diagnosis, Mutation, Prognosis, Young Adult, Chromosome Deletion, Chromosomes, Human, Pair 17 genetics, Multiple Myeloma genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Despite tremendous improvements in the outcome of patients with multiple myeloma in the past decade, high-risk patients have not benefited from the approval of novel drugs. The most important prognostic factor is the loss of parts of the short arm of chromosome 17, known as deletion 17p (del(17p)). A recent publication (on a small number of patients) suggested that these patients are at very high-risk only if del(17p) is associated with TP53 mutations, the so-called "double-hit" population. To validate this finding, we designed a much larger study on 121 patients presenting del(17p) in > 55% of their plasma cells, and homogeneously treated by an intensive approach. For these 121 patients, we performed deep next generation sequencing targeted on TP53. The outcome was then compared with a large control population (2505 patients lacking del(17p)). Our results confirmed that the "double hit" situation is the worst (median survival = 36 months), but that del(17p) alone also confers a poor outcome compared with the control cohort (median survival = 52.8 months vs 152.2 months, respectively). In conclusion, our study clearly confirms the extremely poor outcome of patients displaying "double hit," but also that del(17p) alone is still a very high-risk feature, confirming its value as a prognostic indicator for poor outcome., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
177. Treatment of relapsed and refractory multiple myeloma: recommendations from the International Myeloma Working Group.
- Author
-
Moreau P, Kumar SK, San Miguel J, Davies F, Zamagni E, Bahlis N, Ludwig H, Mikhael J, Terpos E, Schjesvold F, Martin T, Yong K, Durie BGM, Facon T, Jurczyszyn A, Sidana S, Raje N, van de Donk N, Lonial S, Cavo M, Kristinsson SY, Lentzsch S, Hajek R, Anderson KC, João C, Einsele H, Sonneveld P, Engelhardt M, Fonseca R, Vangsted A, Weisel K, Baz R, Hungria V, Berdeja JG, Leal da Costa F, Maiolino A, Waage A, Vesole DH, Ocio EM, Quach H, Driessen C, Bladé J, Leleu X, Riva E, Bergsagel PL, Hou J, Chng WJ, Mellqvist UH, Dytfeld D, Harousseau JL, Goldschmidt H, Laubach J, Munshi NC, Gay F, Beksac M, Costa LJ, Kaiser M, Hari P, Boccadoro M, Usmani SZ, Zweegman S, Holstein S, Sezer O, Harrison S, Nahi H, Cook G, Mateos MV, Rajkumar SV, Dimopoulos MA, and Richardson PG
- Subjects
- Humans, Multiple Myeloma pathology, Neoplasm Recurrence, Local pathology, Antineoplastic Agents therapeutic use, Drug Resistance, Neoplasm drug effects, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Practice Guidelines as Topic standards, Salvage Therapy
- Abstract
This Policy Review presents the International Myeloma Working Group's clinical practice recommendations for the treatment of relapsed and refractory multiple myeloma. Based on the results of phase 2 and phase 3 trials, these recommendations are proposed for the treatment of patients with relapsed and refractory disease who have received one previous line of therapy, and for patients with relapsed and refractory multiple myeloma who have received two or more previous lines of therapy. These recommendations integrate the issue of drug access in both low-income and middle-income countries and in high-income countries to help guide real-world practice and thus improve patient outcomes., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
178. Multiple myeloma: EHA-ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up † .
- Author
-
Dimopoulos MA, Moreau P, Terpos E, Mateos MV, Zweegman S, Cook G, Delforge M, Hájek R, Schjesvold F, Cavo M, Goldschmidt H, Facon T, Einsele H, Boccadoro M, San-Miguel J, Sonneveld P, and Mey U
- Subjects
- Follow-Up Studies, Humans, Societies, Medical, Multiple Myeloma diagnosis, Multiple Myeloma epidemiology, Multiple Myeloma therapy
- Abstract
Competing Interests: Disclosure MAD reported consultancy and honoraria from Janssen, Celgene, Takeda, Amgen and Bristol Myers Squibb; PM reported honoraria from Celgene, Janssen, Takeda, Amgen and Abbvie; ET reported honoraria from Bristol Myers Squibb, Janssen, Celgene, Takeda, Genesis Pharma, Amgen, Sanofi and Novartis; research funding from Janssen, Amgen, Takeda, Sanofi and Genesis Pharma; MVM reported honoraria from lectures and boards from Janssen, Celgene, Amgen, Takeda, Abbvie, GlaxoSmithKline, Adaptive, Roche and Seattle Genetics; SZ reported participation in advisory boards for Takeda, Celgene, Janssen, Sanofi and Oncopeptides and research funding from Celgene, Janssen and Takeda; GC reported being a member of speaker bureau for Takeda, Bristol Myers Squibb, Celgene, Amgen, Sanofi and Janssen and has received research grants from Bristol Myers Squibb, Celgene and Takeda; MD reported honoraria from Abbvie, Adaptive Biotechnologies, Amgen, Bristol Myers Squibb, Celgene, Janssen, Karyopharm, Sanofi and Takeda and has received research funding from Bristol Myers Squibb, Celgene, Janssen and Takeda; RH reported consultant or advisory roles for Janssen, Amgen, Celgene, AbbVie, Bristol Myers Squibb, Novartis, PharmaMar and Takeda; honoraria from Janssen, Amgen, Celgene, Bristol Myers Squibb, PharmaMar and Takeda and has received research grants from Janssen, Amgen, Celgene, Bristol Myers Squibb, Novartis and Takeda; FS reported honoraria from Amgen, Celgene, Bristol Myers Squibb, Takeda, Abbvie, Janssen, Novartis, SkyliteDX, Oncopeptides, Sanofi, GlaxoSmithKline, Adaptive and Merck Sharp & Dohme; MC reported honoraria from Janssen, Celgene, Amgen, Bristol Myers Squibb, Takeda, AbbVie, Sanofi and Adaptive Biotechnologies and speaker's bureau membership for Janssen and Celgene; HG reported grants from Amgen, Bristol Myers Squibb, Celgene, Chugai, Dietmar-Hopp-Foundation, Janssen, John Hopkins University and Sanofi; research support from Amgen, Bristol Myers Squibb, Celgene, Chugai, Janssen, Incyte, Molecular Partners, Merck Sharp & Dohme, Sanofi, Mundipharma, Takeda and Novartis; participation in advisory boards for Adaptive Biotechnology, Amgen, Bristol Myers Squibb, Celgene, Janssen, Sanofi and Takeda; honoraria from Academy2 GmbH & Co. KG, Amgen, ArtTempi, Bristol Myers Squibb, Celgene, Chop GmbH, Chugai, FomF GmbH, GlaxoSmithKline, GWT Forschung und Innovation Dresden, InVo Institut für Versorgungsforschung in der Onkologie GbR, Janssen, Kompetenznetz Maligne Lymphome (KML), MedConcept GmbH, Medical Communication GmbH, New Concept Oncology, Novartis, Omnia Med Deutschland and Sanofi; TF reported speaker and advisory roles for Janssen, Bristol Myers Squibb and Takeda; role for Roche, Sanofi, Karyopharm and Oncopeptides and speaker role for Amgen; HE reported consulting and advisory roles for Bristol Myers Squibb, Celgene, Janssen, Amgen, Takeda, Sanofi and GlaxoSmithKline; research funding from Bristol Myers Squibb, Celgene, Janssen, Amgen, GlaxoSmithKline and Sanofi; honoraria from Bristol Myers Squibb, Celgene, Amgen, Takeda, Sanofi and GlaxoSmithKline; MB has received honoraria from Sanofi, Celgene, Amgen, Janssen, Novartis, Bristol Myers Squibb and AbbVie; has served on advisory boards for Janssen and GlaxoSmithKline; has received research funding from Sanofi, Celgene, Amgen, Janssen, Novartis, Bristol Myers Squibb and Mundipharma; JS-M reported consultancy for Amgen, Bristol Myers Squibb, Celgene, Janssen, Merck Sharp & Dohme, Novartis, GlaxoSmithKline, Takeda, Sanofi and Roche; PS reported honoraria and advisory roles for Celgene, Janssen, Amgen, Takeda, Bristol Myers Squibb and Skyline and research funding from Celgene, Amgen, Janssen and Takeda; UM reported honoraria from Celgene, Janssen, Amgen, Takeda, AbbVie, Bristol Myers Squibb and Sanofi.
- Published
- 2021
- Full Text
- View/download PDF
179. Front-line daratumumab-VTd versus standard-of-care in ASCT-eligible multiple myeloma: matching-adjusted indirect comparison.
- Author
-
Moreau P, Hebraud B, Facon T, Leleu X, Hulin C, Hashim M, Hu Y, Caillot D, Benboubker L, Zweegman S, Merz M, Weisel K, Salwender H, Mai EK, Goldschmidt H, Bertsch U, Vanquickelberghe V, Kampfenkel T, Boer C, Krotneva S, Proskorovsky I, He J, Lam A, Lee C, Cote S, and Sonneveld P
- Subjects
- Adult, Aged, Bortezomib therapeutic use, Cyclophosphamide therapeutic use, Dexamethasone therapeutic use, Female, Humans, Induction Chemotherapy, Male, Middle Aged, Multiple Myeloma mortality, Multiple Myeloma pathology, Multiple Myeloma surgery, Progression-Free Survival, Randomized Controlled Trials as Topic, Stem Cell Transplantation, Survival Rate, Thalidomide therapeutic use, Transplantation, Autologous, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma drug therapy
- Abstract
Aim: To compare daratumumab plus standard-of-care (SoC; bortezomib/thalidomide/dexamethasone [VTd]) and VTd alone with other SoC for transplant-eligible newly diagnosed multiple myeloma. Patients & methods: We conducted an unanchored matching-adjusted indirect comparison of progression-free and overall survival (PFS/OS) with D-VTd/VTd versus bortezomib/lenalidomide/dexamethasone (VRd), bortezomib/cyclophosphamide/dexamethasone (VCd) and bortezomib/dexamethasone (Vd). Results: After matching adjustment, significant improvements in PFS were estimated for D-VTd versus VRd (hazard ratio [HR]: 0.47 [95% CI: 0.33-0.69]), VCd (HR: 0.35 [95% CI: 0.21-0.58]) and Vd (HR: 0.42 [95% CI: 0.28-0.63]). OS was significantly longer with D-VTd versus VRd (HR: 0.31 [95% CI: 0.16-0.57]), VCd (HR: 0.35 [95% CI: 0.14-0.86]) and Vd (HR: 0.38 [95% CI: 0.18-0.77]). No significant PFS/OS differences were seen for VTd versus other SoC. Conclusion: This analysis supports front-line daratumumab for transplant-eligible newly diagnosed multiple myeloma.
- Published
- 2021
- Full Text
- View/download PDF
180. Health-Related Quality of Life in Transplant-Ineligible Patients With Newly Diagnosed Multiple Myeloma: Findings From the Phase III MAIA Trial.
- Author
-
Perrot A, Facon T, Plesner T, Usmani SZ, Kumar S, Bahlis NJ, Hulin C, Orlowski RZ, Nahi H, Mollee P, Ramasamy K, Roussel M, Jaccard A, Delforge M, Karlin L, Arnulf B, Chari A, He J, Ho KF, Van Rampelbergh R, Uhlar CM, Wang J, Kobos R, Gries KS, Fastenau J, and Weisel K
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone adverse effects, Female, Humans, Lenalidomide adverse effects, Male, Middle Aged, Multiple Myeloma diagnosis, Pain Measurement, Time Factors, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Dexamethasone therapeutic use, Lenalidomide therapeutic use, Multiple Myeloma drug therapy, Patient Reported Outcome Measures, Quality of Life
- Abstract
Purpose: To evaluate the effects of daratumumab, lenalidomide, and dexamethasone (D-Rd) versus lenalidomide and dexamethasone (Rd) on patient-reported outcomes (PROs) in the phase III MAIA study., Patients and Methods: PROs were assessed on the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30-item and the EuroQol 5-dimensional descriptive system at baseline and every 3 months during treatment. By mixed-effects model, changes from baseline are presented as least squares means with 95% CIs., Results: A total of 737 transplant-ineligible (TIE) patients with newly diagnosed multiple myeloma were randomly assigned to D-Rd (n = 368) or Rd (n = 369). Compliance with PRO assessments was high at baseline (> 90%) through month 12 (> 78%) for both groups. European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30-item global health status scores improved from baseline in both groups and were consistently greater with D-Rd at all time points. A global health status benefit was achieved with D-Rd, regardless of age (< 75 and ≥ 75 years), baseline Eastern Cooperative Oncology Group (ECOG) performance status score, or depth of response. D-Rd treatment resulted in significantly greater reduction in pain scores as early as cycle 3 ( P = .0007 v Rd); the magnitude of change was sustained through cycle 12. Reductions in pain with D-Rd were clinically meaningful in patients regardless of age, ECOG status, or depth of response. Similarly, PRO improvements were observed with D-Rd and Rd on the EuroQol 5-dimensional descriptive system visual analog scale score., Conclusion: D-Rd compared with Rd was associated with faster and sustained clinically meaningful improvements in PROs, including pain, in transplant-ineligible patients with newly diagnosed multiple myeloma regardless of age, baseline ECOG status, or depth of treatment response.
- Published
- 2021
- Full Text
- View/download PDF
181. The molecular make up of smoldering myeloma highlights the evolutionary pathways leading to multiple myeloma.
- Author
-
Boyle EM, Deshpande S, Tytarenko R, Ashby C, Wang Y, Bauer MA, Johnson SK, Wardell CP, Thanendrarajan S, Zangari M, Facon T, Dumontet C, Barlogie B, Arbini A, Rustad EH, Maura F, Landgren O, Zhan F, van Rhee F, Schinke C, Davies FE, Morgan GJ, and Walker BA
- Subjects
- APOBEC Deaminases genetics, Adult, Aged, Aged, 80 and over, Alleles, Biomarkers, Tumor metabolism, Clone Cells, DNA Copy Number Variations genetics, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Genome, Human, Humans, Male, Middle Aged, Multivariate Analysis, Mutation genetics, Mutation Rate, Progression-Free Survival, Proto-Oncogene Proteins p21(ras) genetics, Smoldering Multiple Myeloma diagnosis, Time Factors, Translocation, Genetic, Evolution, Molecular, Smoldering Multiple Myeloma genetics
- Abstract
Smoldering myeloma (SMM) is associated with a high-risk of progression to myeloma (MM). We report the results of a study of 82 patients with both targeted sequencing that included a capture of the immunoglobulin and MYC regions. By comparing these results to newly diagnosed myeloma (MM) we show fewer NRAS and FAM46C mutations together with fewer adverse translocations, del(1p), del(14q), del(16q), and del(17p) in SMM consistent with their role as drivers of the transition to MM. KRAS mutations are associated with a shorter time to progression (HR 3.5 (1.5-8.1), p = 0.001). In an analysis of change in clonal structure over time we studied 53 samples from nine patients at multiple time points. Branching evolutionary patterns, novel mutations, biallelic hits in crucial tumour suppressor genes, and segmental copy number changes are key mechanisms underlying the transition to MM, which can precede progression and be used to guide early intervention strategies.
- Published
- 2021
- Full Text
- View/download PDF
182. Recommendations for vaccination in multiple myeloma: a consensus of the European Myeloma Network.
- Author
-
Ludwig H, Boccadoro M, Moreau P, San-Miguel J, Cavo M, Pawlyn C, Zweegman S, Facon T, Driessen C, Hajek R, Dimopoulos MA, Gay F, Avet-Loiseau H, Terpos E, Zojer N, Mohty M, Mateos MV, Einsele H, Delforge M, Caers J, Weisel K, Jackson G, Garderet L, Engelhardt M, van de Donk N, Leleu X, Goldschmidt H, Beksac M, Nijhof I, Abildgaard N, Bringhen S, and Sonneveld P
- Subjects
- Clinical Trials as Topic, Consensus, Europe, Humans, Immunosuppressive Agents administration & dosage, Immunosuppressive Agents adverse effects, Immunosuppressive Agents therapeutic use, Multiple Myeloma immunology, Multiple Myeloma therapy, Communicable Disease Control, Communicable Diseases etiology, Multiple Myeloma complications, Practice Guidelines as Topic, Vaccination, Vaccines administration & dosage
- Abstract
Vaccination is one of the most successful medical interventions that has saved the life of millions of people. Vaccination is particularly important in patients with multiple myeloma, who have an increased risk of infections due to the disease-inherent immune suppression, and because of the immune suppressive effects of therapy. Hence, all appropriate measures should be exploited, to elicit an effective immune response to common pathogens like influenza, pneumococci, varicella zoster virus, and to those bacteria and viruses (haemophilus influenzae, meningococci, and hepatitis) that frequently may pose a significant risk to patients with multiple myeloma. Patients after autologous, and specifically after allogeneic transplantation have severely reduced antibody titers, and therefore require a broader spectrum of vaccinations. Response to vaccination in myeloma often is less vigorous than in the general population, mandating either measurement of the postvaccination antibody titers and/or repeating the vaccination. Here, we compile the existing data on vaccination in multiple myeloma and provide recommendations for clinical practice.
- Published
- 2021
- Full Text
- View/download PDF
183. International harmonization in performing and reporting minimal residual disease assessment in multiple myeloma trials.
- Author
-
Costa LJ, Derman BA, Bal S, Sidana S, Chhabra S, Silbermann R, Ye JC, Cook G, Cornell RF, Holstein SA, Shi Q, Omel J, Callander NS, Chng WJ, Hungria V, Maiolino A, Stadtmauer E, Giralt S, Pasquini M, Jakubowiak AJ, Morgan GJ, Krishnan A, Jackson GH, Mohty M, Mateos MV, Dimopoulos MA, Facon T, Spencer A, Miguel JS, Hari P, Usmani SZ, Manier S, McCarthy P, Kumar S, Gay F, and Paiva B
- Subjects
- Clinical Trials as Topic, Diagnostic Imaging, Disease Management, Drug Collateral Sensitivity, Global Health, Humans, Molecular Diagnostic Techniques methods, Molecular Diagnostic Techniques standards, Multiple Myeloma therapy, Neoplastic Cells, Circulating metabolism, Neoplastic Cells, Circulating pathology, Outcome Assessment, Health Care, Population Surveillance, Reproducibility of Results, Smoldering Multiple Myeloma epidemiology, Smoldering Multiple Myeloma pathology, Time Factors, Multiple Myeloma epidemiology, Multiple Myeloma pathology, Neoplasm, Residual diagnosis, Neoplasm, Residual epidemiology
- Abstract
Minimal residual disease (MRD) assessment is incorporated in an increasing number of multiple myeloma (MM) clinical trials as a correlative analysis, an endpoint or even as a determinant of subsequent therapy. There is substantial heterogeneity across clinical trials in how MRD is assessed and reported, creating challenges for data interpretation and for the design of subsequent studies. We convened an international panel of MM investigators to harmonize how MRD should be assessed and reported in MM clinical trials. The panel provides consensus on which MM trials should include MRD, the recommended time points for MRD assessment, and expected analytical validation for MRD assays. We subsequently outlined parameters for reporting MRD results implementing the intention-to-treat principle. The panel provides guidance regarding the incorporation of newer peripheral blood-based and imaging-based approaches to detection of residual disease. Recommendations are summarized in 13 consensus statements that should be followed by sponsors, investigators, editors, and reviewers engaged in designing, performing, and interpreting MM trials.
- Published
- 2021
- Full Text
- View/download PDF
184. Continuous lenalidomide and low-dose dexamethasone in patients with transplant-ineligible newly diagnosed MM: FIRST trial subanalysis of Canadian/US patients.
- Author
-
Belch A, Bahlis N, White D, Cheung M, Chen C, Shustik C, Song K, Tosikyan A, Dispenzieri A, Anderson K, Brown D, Robinson S, Srinivasan S, and Facon T
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Canada, Dexamethasone adverse effects, Disease Progression, Drug Administration Schedule, Eligibility Determination, Female, Hematopoietic Stem Cell Transplantation, Humans, Lenalidomide adverse effects, Male, Melphalan administration & dosage, Middle Aged, Multiple Myeloma diagnosis, Multiple Myeloma mortality, Prednisone administration & dosage, Progression-Free Survival, Thalidomide administration & dosage, Time Factors, United States, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Dexamethasone administration & dosage, Lenalidomide administration & dosage, Multiple Myeloma drug therapy
- Abstract
The phase 3 FIRST trial demonstrated significant improvement in progression-free survival (PFS) and overall survival (OS) with an immune-stimulatory agent, lenalidomide, in combination with low-dose dexamethasone until disease progression (Rd continuous) vs melphalan +prednisone + thalidomide (MPT) in transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM). Rd continuous similarly extended PFS vs fixed-duration Rd for 18 cycles (Rd18). Outcomes in the Canadian/US subgroup (104 patients per arm) are reported in this analysis. Rd continuous demonstrated a significant improvement in PFS vs MPT (median, 29.3 vs 20.2 months; HR, 0.69 [95% CI, 0.49-0.97]; p = 0.03326) and an improvement vs Rd18 (median, 21.9 months). Median OS was 56.9 vs 46.8 months with Rd continuous vs MPT (p = 0.15346) and 59.5 months with Rd18. The overall response rate was higher with Rd continuous and Rd18 (78.8% and 79.8%) vs MPT (65.4%). In the 49.0%, 52.9%, and 29.8% of patients with at least very good partial response in the Rd continuous, Rd18, and MPT arms, respectively, the median PFS was 56.0, 30.9, and 40.2 months, respectively. The most common grade 3/4 treatment-emergent adverse events were neutropenia (28.4%, 30.1%, and 52.0%), anemia (23.5%, 21.4%, and 23.5%), and infections (37.3%, 30.1%, and 24.5%) with Rd continuous, Rd18, and MPT, respectively. These results were consistent with those in the intent-to-treat population, confirming the benefit of Rd continuous vs MPT in the Canadian/US subgroup and supporting the role of Rd continuous as a standard of care for transplant-ineligible patients with NDMM., (© 2020 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
185. Daratumumab-lenalidomide-dexamethasone vs standard-of-care regimens: Efficacy in transplant-ineligible untreated myeloma.
- Author
-
Durie BGM, Kumar SK, Usmani SZ, Nonyane BAS, Ammann EM, Lam A, Kobos R, Maiese EM, and Facon T
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone administration & dosage, Dexamethasone adverse effects, Disease-Free Survival, Female, Humans, Lenalidomide administration & dosage, Lenalidomide adverse effects, Male, Survival Rate, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Multiple Myeloma drug therapy, Multiple Myeloma mortality
- Abstract
Daratumumab in combination with lenalidomide-dexamethasone (D-Rd) recently received FDA approval for the treatment of transplant-ineligible patients with newly diagnosed multiple myeloma (NDMM). The present PEGASUS study compared progression-free survival (PFS) in patients treated with D-Rd in the MAIA trial and patients treated with common standard-of-care regimens from the Flatiron Health electronic health record-derived deidentified database, which has data from patients treated primarily at community-based oncology practices in the United States. Individual-level patient data from both data sources were used to perform an anchored indirect treatment comparison (ITC) of D-Rd to bortezomib-lenalidomide-dexamethasone (VRd) and bortezomib-dexamethasone (Vd); lenalidomide-dexamethasone (Rd) was the common anchor for the ITC. Hazard ratios (HRs) reflecting direct comparisons of PFS within MAIA (D-Rd vs Rd) and Flatiron Health (VRd vs Rd; Vd vs Rd) were used to make ITCs for D-Rd vs VRd and Vd, respectively. After application of MAIA inclusion/exclusion criteria and propensity-score weighting, the Flatiron Health patients resembled the MAIA trial population on measured baseline characteristics. Based on the direct comparison within MAIA, treatment with D-Rd was associated with a significantly lower risk of progression or death compared to Rd (HR 0.54; 95% CI 0.42, 0.71). Based on the ITCs, D-Rd was associated with a significantly lower risk of progression or death compared to VRd (HR 0.68; 95% CI 0.48, 0.98) and Vd (HR 0.48; 95% CI 0.33, 0.69). In the absence of head-to-head trials comparing D-Rd to VRd or Vd, the present ITC may help inform treatment selection in transplant-ineligible patients with NDMM., (© 2020 The Authors. American Journal of Hematology published by Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
186. Once-per-week selinexor, bortezomib, and dexamethasone versus twice-per-week bortezomib and dexamethasone in patients with multiple myeloma (BOSTON): a randomised, open-label, phase 3 trial.
- Author
-
Grosicki S, Simonova M, Spicka I, Pour L, Kriachok I, Gavriatopoulou M, Pylypenko H, Auner HW, Leleu X, Doronin V, Usenko G, Bahlis NJ, Hajek R, Benjamin R, Dolai TK, Sinha DK, Venner CP, Garg M, Gironella M, Jurczyszyn A, Robak P, Galli M, Wallington-Beddoe C, Radinoff A, Salogub G, Stevens DA, Basu S, Liberati AM, Quach H, Goranova-Marinova VS, Bila J, Katodritou E, Oliynyk H, Korenkova S, Kumar J, Jagannath S, Moreau P, Levy M, White D, Gatt ME, Facon T, Mateos MV, Cavo M, Reece D, Anderson LD Jr, Saint-Martin JR, Jeha J, Joshi AA, Chai Y, Li L, Peddagali V, Arazy M, Shah J, Shacham S, Kauffman MG, Dimopoulos MA, Richardson PG, and Delimpasi S
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib adverse effects, Dexamethasone adverse effects, Drug Administration Schedule, Female, Humans, Hydrazines adverse effects, Kaplan-Meier Estimate, Male, Middle Aged, Progression-Free Survival, Triazoles adverse effects, Antineoplastic Agents administration & dosage, Bortezomib administration & dosage, Dexamethasone administration & dosage, Hydrazines administration & dosage, Multiple Myeloma drug therapy, Triazoles administration & dosage
- Abstract
Background: Selinexor combined with dexamethasone has shown activity in patients with heavily pre-treated multiple myeloma. In a phase 1b/2 study, the combination of oral selinexor with bortezomib (a proteasome inhibitor) and dexamethasone induced high response rates with low rates of peripheral neuropathy, the main dose-limiting toxicity of bortezomib. We aimed to evaluate the clinical benefit of weekly selinexor, bortezomib, and dexamethasone versus standard bortezomib and dexamethasone in patients with previously treated multiple myeloma., Methods: This phase 3, randomised, open-label trial was done at 123 sites in 21 countries. Patients aged 18 years or older, who had multiple myeloma, and who had previously been treated with one to three lines of therapy, including proteasome inhibitors, were randomly allocated (1:1) to receive selinexor (100 mg once per week), bortezomib (1·3 mg/m
2 once per week), and dexamethasone (20 mg twice per week), or bortezomib (1·3 mg/m2 twice per week for the first 24 weeks and once per week thereafter) and dexamethasone (20 mg four times per week for the first 24 weeks and twice per week thereafter). Randomisation was done using interactive response technology and stratified by previous proteasome inhibitor therapy, lines of treatment, and multiple myeloma stage. The primary endpoint was progression-free survival in the intention-to-treat population. Patients who received at least one dose of study treatment were included in the safety population. This trial is registered at ClinicalTrials.gov, NCT03110562. The trial is ongoing, with 55 patients remaining on randomised therapy as of Feb 20, 2020., Findings: Of 457 patients screened for eligibility, 402 were randomly allocated-195 (49%) to the selinexor, bortezomib, and dexamethasone group and 207 (51%) to the bortezomib and dexamethasone group-and the first dose of study medication was given between June 6, 2017, and Feb 5, 2019. Median follow-up durations were 13·2 months [IQR 6·2-19·8] for the selinexor, bortezomib, and dexamethasone group and 16·5 months [9·4-19·8] for the bortezomib and dexamethasone group. Median progression-free survival was 13·93 months (95% CI 11·73-not evaluable) with selinexor, bortezomib, and dexamethasone and 9·46 months (8·11-10·78) with bortezomib and dexamethasone (hazard ratio 0·70 [95% CI 0·53-0·93], p=0·0075). The most frequent grade 3-4 adverse events were thrombocytopenia (77 [39%] of 195 patients in the selinexor, bortezomib, and dexamethasone group vs 35 [17%] of 204 in the bortezomib and dexamethasone group), fatigue (26 [13%] vs two [1%]), anaemia (31 [16%] vs 20 [10%]), and pneumonia (22 [11%] vs 22 [11%]). Peripheral neuropathy of grade 2 or above was less frequent with selinexor, bortezomib, and dexamethasone (41 [21%] patients) than with bortezomib and dexamethasone (70 [34%] patients; odds ratio 0·50 [95% CI 0·32-0·79], p=0·0013). 47 (24%) patients in the selinexor, bortezomib, and dexamethasone group and 62 (30%) in the bortezomib and dexamethasone group died., Interpretation: A once-per-week regimen of selinexor, bortezomib, and dexamethasone is a novel, effective, and convenient treatment option for patients with multiple myeloma who have received one to three previous lines of therapy., Funding: Karyopharm Therapeutics., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
187. Efficacy and safety of carfilzomib-based regimens in frail patients with relapsed and/or refractory multiple myeloma.
- Author
-
Facon T, Niesvizky R, Mateos MV, Siegel D, Rosenbaum C, Bringhen S, Weisel K, Ho PJ, Ludwig H, Kumar S, Wang K, Obreja M, Yang Z, Klippel Z, Mezzi K, Goldrick A, Tekle C, and Dimopoulos MA
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Frail Elderly, Humans, Oligopeptides adverse effects, Multiple Myeloma drug therapy
- Abstract
Frailty is most prevalent among elderly multiple myeloma (MM) patients, and frail patients have a higher risk of poor outcomes due to reduced performance status or comorbidities. This post hoc analysis assessed efficacy and safety of carfilzomib combinations in frail patients with relapsed and/or refractory MM from the phase 3 ASPIRE (carfilzomib [27 mg/m2]-lenalidomide-dexamethasone [KRd27] vs lenalidomide-dexamethasone [Rd]), ENDEAVOR (carfilzomib [56 mg/m2]-dexamethasone [Kd56] vs bortezomib-dexamethasone [Vd]), and ARROW (once-weekly carfilzomib [70 mg/m2]-dexamethasone [Kd70] vs carfilzomib [27 mg/m2]-dexamethasone [Kd27]) studies. A frailty algorithm incorporating age, Charlson comorbidity index, and performance status classified patients as fit, intermediate, or frail. Results are presented for frail patients (ASPIRE, n = 196; ENDEAVOR, n = 330; ARROW, n = 141). In ASPIRE, median progression-free survival (PFS) (hazard ratio; 95% confidence interval) was 24.1 (KRd27) vs 15.9 months (Rd) (0.78; 0.54-1.12); median overall survival (OS) was 36.4 vs 26.2 months (0.79; 0.57-1.08). In ENDEAVOR, median PFS was 18.7 (Kd56) vs 6.6 months (Vd) (0.50; 0.36-0.68); median OS was 33.6 vs 21.8 months (0.75; 0.56-1.00). In ARROW, median PFS was 10.3 (once-weekly Kd70) vs 6.6 months (twice-weekly Kd27) (0.76; 0.49-1.16). In all 3 studies, rates of grade ≥3 treatment-emergent adverse events were consistent with those observed in the primary studies. The ASPIRE, ENDEAVOR, and ARROW primary analyses demonstrated favorable benefit-risk profiles with carfilzomib-containing regimens compared with controls. Across clinically relevant subgroups, including those by frailty status, consistent efficacy and safety were observed with KRd27, Kd56, and weekly Kd70, and treatment with these regimens should not be restricted by frailty status., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
188. Evaluation of Daratumumab for the Treatment of Multiple Myeloma in Patients With High-risk Cytogenetic Factors: A Systematic Review and Meta-analysis.
- Author
-
Giri S, Grimshaw A, Bal S, Godby K, Kharel P, Djulbegovic B, Dimopoulos MA, Facon T, Usmani SZ, Mateos MV, and Costa LJ
- Subjects
- Antibodies, Monoclonal adverse effects, Cytogenetic Analysis, Humans, Progression-Free Survival, Multiple Myeloma drug therapy, Multiple Myeloma genetics
- Abstract
Importance: The addition of daratumumab to backbone multiple myeloma (MM) regimens is associated with improved response rates and progression-free survival (PFS). Whether improved outcomes are also associated with this regimen among patients with cytogenetically defined high-risk MM (HRMM) remains unclear., Objective: To measure PFS associated with adding daratumumab to backbone MM regimens among patients with HRMM., Data Sources: For this systematic review and meta-analysis, MEDLINE, Embase, PubMed, Scopus, Web of Science Core Collection, Cochrane Library, clinical trials registries, and meeting libraries were searched from inception to January 2, 2020, using terms reflecting multiple myeloma and daratumumab., Study Selection: Included studies were phase 3 randomized clinical trials that compared backbone MM regimens with the same regimen plus daratumumab in newly diagnosed or relapsed or refractory MM, such that the only difference between the intervention and control groups was use of daratumumab and reported outcomes by cytogenetic risk. High-risk MM was defined as the presence of t(4;14), t(14;16), or del(17p)., Data Extraction and Synthesis: Using the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guideline, 2 investigators independently extracted study data, with disagreements resolved by a third investigator. Quality was assessed by the Cochrane risk-of-bias method., Main Outcomes and Measures: Data on effectiveness were extracted using hazard ratios (HRs) for PFS. Relative log-HRs were pooled using a DerSimonian-Laird random-effects model. Heterogeneity was assessed using the Cochran Q and the I2 statistic., Results: Of 5194 studies screened, 6 phase 3 trials were eligible, including 3 trials for newly diagnosed MM (2528 patients; 358 with HRMM) and 3 trials for relapsed or refractory MM (1533 patients; 222 with HRMM). Among patients with newly diagnosed HRMM, the addition of daratumumab to backbone regimens was associated with improved PFS (pooled HR, 0.67; 95% CI, 0.47-0.95; P = .02), with little evidence of heterogeneity (Cochran Q, P = .77; I2 = 0%). Similar results were seen among patients with relapsed or refractory HRMM (pooled HR, 0.45; 95% CI, 0.30-0.67; P < .001), again with little evidence of heterogeneity (Cochran Q, P = .63; I2 = 0%)., Conclusions and Relevance: This study suggests that incorporating daratumumab to backbone regimens may be associated with improved PFS among patients with newly diagnosed HRMM or relapsed or refractory HRMM.
- Published
- 2020
- Full Text
- View/download PDF
189. Defining the vulnerable patient with myeloma-a frailty position paper of the European Myeloma Network.
- Author
-
Cook G, Larocca A, Facon T, Zweegman S, and Engelhardt M
- Subjects
- Aged, Aged, 80 and over, Biomarkers, Tumor metabolism, Frail Elderly, Humans, Multiple Myeloma metabolism, Multiple Myeloma therapy, Precision Medicine, Frailty, Multiple Myeloma pathology
- Abstract
As the treatment landscape continues to evolve towards the application of precision medicine in multiple myeloma (MM), there is a clear need to identify those patients who are at risk of not achieving the maximum benefit whilst exposed to the highest level of toxicity. This group of patients, defined as frail, is an unmet clinical need. However, how we define such a vulnerable group of patients with MM remains to be clarified. An integral aspect of this is to define the physiological age and capacity of patients with MM to deal with the burden of their disease and it's treatment. Such assessments may include not only functional and clinical assessments but also laboratory-based biomarkers of frailty, aging and senescent cellular burden. A need to develop, test and validate clinical screening scores before their adoption into clinical practice is mandated. This position paper from the European Myeloma Network aims to review what is known about defining frailty in MM, and how we can advance this knowledge for the design of clinical trials and ultimately how we deliver treatment in the clinic.
- Published
- 2020
- Full Text
- View/download PDF
190. Early relapse after autologous transplant for myeloma is associated with poor survival regardless of cytogenetic risk.
- Author
-
Corre J, Montes L, Martin E, Perrot A, Caillot D, Leleu X, Belhadj K, Facon T, Hulin C, Mohty M, Fontan J, Macro M, Brechignac S, Jaccard A, Stoppa AM, Orsini-Piocelle F, Adiko D, Voillat L, Keddar F, Barry M, Demarquette H, Certain MN, Plantier I, Roussel M, Hébraud B, Filleron T, Attal M, and Avet-Loiseau H
- Subjects
- Autografts, Cytogenetic Analysis, Disease-Free Survival, Humans, Neoplasm Recurrence, Local, Transplantation, Autologous, Hematopoietic Stem Cell Transplantation adverse effects, Multiple Myeloma genetics, Multiple Myeloma therapy
- Published
- 2020
- Full Text
- View/download PDF
191. Matching-adjusted indirect comparison of efficacy and safety of bortezomib, thalidomide, and dexamethasone (VTd) as per label compared with modified VTd dosing schedules in patients with newly diagnosed multiple myeloma who are transplant eligible.
- Author
-
Sonneveld P, Mateos MV, Alegre A, Facon T, Hulin C, Hashim M, Vincken T, Kampfenkel T, Cote S, He J, Lam A, and Moreau P
- Abstract
Background: The combination of bortezomib, thalidomide, and dexamethasone (VTd) is a standard of care for transplant-eligible patients with newly diagnosed multiple myeloma (NDMM). Although approved labeling for VTd includes an escalating thalidomide dose up to 200 mg daily (VTd-label), a lower fixed dose of thalidomide (100 mg daily; VTd-mod) has become commonplace in clinical practice. To date, no clinical trials comparing VTd-mod with VTd-label have been performed. Here, we compared outcomes for VTd-mod with VTd-label using a matching-adjusted indirect comparison., Methods: VTd-mod data were from NCT02541383 (CASSIOPEIA; phase III) and NCT00531453 (phase II); VTd-label data were from NCT00461747 (PETHEMA/GEM; phase III). To adjust for heterogeneity, baseline characteristics from VTd-label were weighted to match VTd-mod. Outcomes included overall survival (OS), progression-free survival (PFS), postinduction and posttransplant responses, and safety., Results: VTd-mod was noninferior to VTd-label for OS, postinduction overall response rate (ORR), and very good partial response or better (≥VGPR). VTd-mod was significantly better than VTd-label for PFS, posttransplant ORR, and ≥VGPR. VTd-mod was noninferior to VTd-label for safety outcomes, and inferior to VTd-label for postinduction and posttransplant complete response or better., Conclusions: Our analysis supports the continued use of VTd-mod in clinical practice in transplant-eligible NDMM patients., Competing Interests: PS: Honoraria from Amgen, BMS, Celgene, Janssen, Karyopharm; Takeda; research funding from Amgen, Celgene, Janssen, Karyopharm, SkylineDx, Takeda. M‐VM: advisory committee for AbbVie, Amgen, Celgene, Genentech, GSK, Janssen, Mundipharma EDO, PharmaMar, Roche Laboratories, Inc, Takeda; membership on an entity's board of directors for AbbVie, Amgen, Celgene, EDO, GSK, Janssen, PharmaMar, Takeda; honoraria from Adaptive, Amgen, Celgene, Janssen, Takeda; speakers bureau for Amgen, Celgene, Janssen, Takeda; data and monitoring committee for Amgen and Jansen. AA: Advisory committee for Amgen, Celgene, Janssen, Sanofi, Takeda; consultancy for Sanofi; honoraria from Amgen, Celgene, Janssen; research funding from Celgene, Janssen, Sanofi. TF: Membership on an entity's board of directors or advisory committees for Amgen, Celgene, Janssen, Karyopharm, Oncopeptides, Roche, Sanofi, Takeda; speakers bureau for Celgene, Janssen, Takeda. CH: Consultancy for Celgene; honoraria from AbbVie, Amgen, Celgene, Janssen. MH and TV: Employment from Ingress‐Health. TK, SC, JH, and AL: Employment from Janssen. PM: Consultancy for AbbVie, Amgen, Celgene, Janssen, Takeda; honoraria from AbbVie, Amgen, Celgene, Janssen, Takeda., (© 2020 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
192. Plasma Cell Leukemia - Facts and Controversies: More Questions than Answers?
- Author
-
Suska A, Vesole DH, Castillo JJ, Kumar SK, Parameswaran H, Mateos MV, Facon T, Gozzetti A, Mikala G, Szostek M, Mikhael J, Hajek R, Terpos E, and Jurczyszyn A
- Abstract
Plasma cell leukemia (PCL) is an aggressive hematological malignancy characterized by an uncontrolled clonal proliferation of plasma cells (PCs) in the bone marrow and peripheral blood. PCL has been defined by an absolute number of circulating PCs exceeding 2.0 × 10
9 /L and/or >20% PCs in the total leucocyte count. It is classified as primary PCL, which develops de novo , and secondary PCL, occurring at the late and advanced stages of multiple myeloma (MM). Primary and secondary PCL are clinically and biologically two distinct entities. After the diagnosis, treatment should be immediate and should include a proteasome inhibitor and immunomodulator-based combination regimens as induction, followed by stem cell transplantation (SCT) in transplant-eligible individuals who have cleared the peripheral blood of circulating PCs. Due to the rarity of the condition, there have been very few clinical trials. Furthermore, virtually all of the myeloma trials exclude patients with active PCL. The evaluation of response has been defined by the International Myeloma Working Group and consists of both acute leukemia and MM criteria. With conventional chemotherapy, the prognosis of primary PCL has been ominous, with reported overall survival (OS) ranging from 6.8 to 12.6 months. The use of novel agents and autologous SCT appears to be associated with deeper response and an improved survival, although it still remains low. The PCL prognostic index provides a simple score to risk-stratify PCL. The prognosis of secondary PCL is extremely poor, with OS of only 1 month., Competing Interests: The authors declare they have no conflicts of interest., (© 2020 International Academy for Clinical Hematology. Publishing services by Atlantis Press International B.V.)- Published
- 2020
- Full Text
- View/download PDF
193. Health-related quality of life results from the IFM 2009 trial: treatment with lenalidomide, bortezomib, and dexamethasone in transplant-eligible patients with newly diagnosed multiple myeloma.
- Author
-
Roussel M, Hebraud B, Hulin C, Perrot A, Caillot D, Stoppa AM, Macro M, Escoffre M, Arnulf B, Belhadj K, Karlin L, Garderet L, Facon T, Guo S, Weng J, Dhanasiri S, Leleu X, Moreau P, and Attal M
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib therapeutic use, Dexamethasone therapeutic use, Humans, Lenalidomide therapeutic use, Quality of Life, Transplantation, Autologous, Treatment Outcome, Hematopoietic Stem Cell Transplantation, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy
- Abstract
The Intergroupe Francophone du Myelome 2009 trial (NCT01191060) assessed health-related quality of life (HRQoL) in patients with newly diagnosed multiple myeloma (NDMM) receiving lenalidomide/bortezomib/dexamethasone (RVd) induction therapy followed by consolidation therapy with either autologous stem cell transplantation (ASCT) plus RVd (RVd-ASCT) or RVd-alone; both groups then received lenalidomide maintenance therapy for 1 year. Global HRQoL, physical functioning, and role functioning scores significantly improved for both cohorts from baseline to the end of consolidation and were sustained during maintenance and follow-up, with clinically meaningful changes (RVd-alone: p = .0002; RVd-ASCT: p < .001). Similarly, both groups showed clinically meaningful improvements from baseline in fatigue, pain, and disease symptom scores. Side effects of treatment scores remained stable. In the RVd-ASCT group, there was transient worsening in HRQoL immediately after ASCT. These findings suggest that the clinical improvements observed with RVd-based treatment are accompanied by overall improvements in HRQoL for patients with NDMM.
- Published
- 2020
- Full Text
- View/download PDF
194. BRAF and DIS3 Mutations Associate with Adverse Outcome in a Long-term Follow-up of Patients with Multiple Myeloma.
- Author
-
Boyle EM, Ashby C, Tytarenko RG, Deshpande S, Wang H, Wang Y, Rosenthal A, Sawyer J, Tian E, Flynt E, Hoering A, Johnson SK, Rutherford MW, Wardell CP, Bauer MA, Dumontet C, Facon T, Thanendrarajan S, Schinke CD, Zangari M, van Rhee F, Barlogie B, Cairns D, Jackson G, Thakurta A, Davies FE, Morgan GJ, and Walker BA
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, DNA Mutational Analysis, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Myeloma drug therapy, Multiple Myeloma mortality, Multiple Myeloma pathology, Prognosis, Survival Rate, Biomarkers, Tumor genetics, Exosome Multienzyme Ribonuclease Complex genetics, Multiple Myeloma genetics, Mutation, Proto-Oncogene Proteins B-raf genetics
- Abstract
Purpose: Copy-number changes and translocations have been studied extensively in many datasets with long-term follow-up. The impact of mutations remains debated given the short time to follow-up of most datasets., Experimental Design: We performed targeted panel sequencing covering 125 myeloma-specific genes and the loci involved in translocations in 223 newly diagnosed myeloma samples recruited into one of the total therapy trials., Results: As expected, the most commonly mutated genes were NRAS, KRAS , and BRAF , making up 44% of patients. Double-Hit and BRAF and DIS3 mutations had an impact on outcome alongside classical risk factors in the context of an intensive treatment approach. We were able to identify both V600E and non-V600E BRAF mutations, 58% of which were predicted to be hypoactive or kinase dead. Interestingly, 44% of the hypoactive/kinase dead BRAF -mutated patients showed co-occurring alterations in KRAS, NRAS , or activating BRAF mutations, suggesting that they play a role in the oncogenesis of multiple myeloma by facilitating MAPK activation and may lead to chemoresistance., Conclusions: Overall, these data highlight the importance of mutational screening to better understand newly diagnosed multiple myeloma and may lead to patient-specific mutation-driven treatment approaches., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
195. Once-weekly (70 mg/m 2 ) vs twice-weekly (56 mg/m 2 ) dosing of carfilzomib in patients with relapsed or refractory multiple myeloma: A post hoc analysis of the ENDEAVOR, A.R.R.O.W., and CHAMPION-1 trials.
- Author
-
Moreau P, Stewart KA, Dimopoulos M, Siegel D, Facon T, Berenson J, Raje N, Berdeja JG, Orlowski RZ, Yang H, Ma H, Klippel Z, Zahlten-Kumeli A, Mezzi K, Iskander K, and Mateos MV
- Subjects
- Aged, Clinical Trials, Phase III as Topic, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multicenter Studies as Topic, Multiple Myeloma pathology, Neoplasm Recurrence, Local pathology, Prognosis, Randomized Controlled Trials as Topic, Salvage Therapy, Survival Rate, Drug Resistance, Neoplasm drug effects, Multiple Myeloma drug therapy, Neoplasm Recurrence, Local drug therapy, Oligopeptides therapeutic use
- Abstract
Combination of carfilzomib with dexamethasone (Kd) is approved for use in relapsed and/or refractory multiple myeloma (RRMM), with carfilzomib administered twice weekly at 56 mg/m
2 (Kd56 BIW) or once weekly at 70 mg/m2 (Kd70 QW). Post hoc cross-trial comparisons were performed to compare efficacy and safety profiles of Kd70 QW vs Kd56 BIW dosing schedules using data from three trials of patients with RRMM: A.R.R.O.W., CHAMPION-1, and ENDEAVOR. To select for comparable patient populations, side-by-side efficacy and safety comparisons were performed in subgroups of patients with 2-3 prior lines of therapy who were not refractory to bortezomib. The overall response rate (ORR) was 69.9% (95% confidence interval [CI], 61.7-77.2) for Kd70 QW and 72.4% (95% CI, 65.9-78.2) for Kd56 BIW. Median progression-free survival (PFS) was 12.1 months (95% CI, 8.4-14.3) for Kd70 QW and 14.5 months (95% CI, 10.2-not evaluable) for Kd56 BIW. Frequency of grade ≥ 3 adverse events (AEs) was 67.6% for Kd70 QW and 85.3% for Kd56 BIW. Regression analyses (adjusting for prognostic factors) of all patients in the trials who received Kd70 QW vs Kd56 BIW estimated a PFS hazard ratio of 0.91 (95% CI, 0.69-1.19; P = .47) and an ORR odds ratio of 1.12 (95% CI, 0.74-1.69; P = .61). These results suggest that Kd70 QW has a comparable efficacy profile compared with Kd56 BIW and represents a convenient and well-tolerated treatment for patients with RRMM., (© 2020 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)- Published
- 2020
- Full Text
- View/download PDF
196. Anti-B-Cell Maturation Antigen BiTE Molecule AMG 420 Induces Responses in Multiple Myeloma.
- Author
-
Topp MS, Duell J, Zugmaier G, Attal M, Moreau P, Langer C, Krönke J, Facon T, Salnikov AV, Lesley R, Beutner K, Kalabus J, Rasmussen E, Riemann K, Minella AC, Munzert G, and Einsele H
- Subjects
- Adult, Aged, Antibodies, Bispecific immunology, B-Cell Maturation Antigen immunology, Dose-Response Relationship, Drug, Female, Humans, Male, Middle Aged, Multiple Myeloma immunology, T-Lymphocytes drug effects, T-Lymphocytes immunology, Antibodies, Bispecific administration & dosage, B-Cell Maturation Antigen antagonists & inhibitors, Multiple Myeloma therapy
- Abstract
Purpose: The anti-B-cell maturation antigen BiTE molecule AMG 420 was assessed in patients with relapsed/refractory multiple myeloma., Patients and Methods: In this first-in-human study, up to 10 cycles of AMG 420 were given (4-week infusions/6-week cycles). Patients had progression after ≥ 2 lines of prior therapy and no extramedullary disease. Minimal residual disease (MRD) response was defined as < 1 tumor cell/10
4 bone marrow cells by flow cytometry., Results: Forty-two patients received AMG 420 at 0.2-800 μg/d. Median age was 65 years, and median disease duration was 5.2 years. Median exposure was 1 cycle (range, 1-10 cycles) and 7 cycles (range, 1-10 cycles) for responders. Patients discontinued for disease progression (n = 25), adverse events (AEs; n = 7), death (n = 4), completion of 10 cycles (n = 3), and consent withdrawal (n = 1). Two patients remain on treatment. There were 2 nontreatment-related deaths from AEs, influenza/aspergillosis and adenovirus-related hepatitis. Serious AEs (n = 20; 48%) included infections (n = 14) and polyneuropathy (n = 2); treatment-related serious AEs included 2 grade 3 polyneuropathies and 1 grade 3 edema. There were no grade ≥ 3 CNS toxicities or anti-AMG 420 antibodies. In this study, 800 μg/d was considered to not be tolerable because of 1 instance each of grade 3 cytokine release syndrome and grade 3 polyneuropathy, both of which resolved. The overall response rate was 31% (n = 13 of 42). At the maximum tolerated dose (MTD) of 400 μg/d, the response rate was 70% (n = 7 of 10). Of these, five patients experienced MRD-negative complete responses, and 1 had a partial response, and 1 had a very good partial response; all 7 patients responded during the first cycle, and some responses lasted > 1 year., Conclusion: In this study of AMG 420 in patients with relapsed/refractory multiple myeloma, the response rate was 70%, including 50% MRD-negative complete responses, at 400 μg/d, the MTD for this study.- Published
- 2020
- Full Text
- View/download PDF
197. A matching-adjusted indirect treatment comparison (MAIC) of daratumumab-bortezomib-melphalan-prednisone (D-VMP) versus lenalidomide-dexamethasone continuous (Rd continuous), lenalidomide-dexamethasone 18 months (Rd 18), and melphalan-prednisone-thalidomide (MPT).
- Author
-
Dimopoulos MA, Cavo M, Mateos MV, Facon T, Heeg B, van Beekhuizen S, Gebregergish SB, Nair S, Pisini M, Lam A, and Slavcev M
- Subjects
- Antibodies, Monoclonal therapeutic use, Bortezomib therapeutic use, Dexamethasone therapeutic use, Humans, Lenalidomide therapeutic use, Melphalan therapeutic use, Prednisone therapeutic use, Thalidomide therapeutic use, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma drug therapy
- Abstract
D-VMP is a novel treatment for transplant-ineligible newly diagnosed multiple myeloma (TIE NDMM). D-VMP significantly prolonged PFS versus VMP in the ALCYONE trial. The FIRST trial investigated Rd given in 28-day cycles until disease progression, Rd for 18 cycles, and MPT for 12 cycles for TIE NDMM. As no randomized controlled trials comparing D-VMP to standard-of-care regimens such as those in FIRST are available, an MAIC was performed to assess relative OS and PFS for D-VMP from ALYCONE and Rd continuous, Rd 18, and MPT from FIRST. Individual patient data for D-VMP in ALCYONE were weighted to match aggregated baseline patient characteristics for each arm of FIRST. D-VMP significantly improved OS versus MPT and Rd 18, with a trend favoring D-VMP versus Rd continuous. D-VMP performed significantly better than all FIRST comparators for PFS. This MAIC demonstrates OS and PFS benefits for D-VMP versus Rd continuous, Rd 18, and MPT.
- Published
- 2020
- Full Text
- View/download PDF
198. Belantamab mafodotin for relapsed or refractory multiple myeloma (DREAMM-2): a two-arm, randomised, open-label, phase 2 study.
- Author
-
Lonial S, Lee HC, Badros A, Trudel S, Nooka AK, Chari A, Abdallah AO, Callander N, Lendvai N, Sborov D, Suvannasankha A, Weisel K, Karlin L, Libby E, Arnulf B, Facon T, Hulin C, Kortüm KM, Rodríguez-Otero P, Usmani SZ, Hari P, Baz R, Quach H, Moreau P, Voorhees PM, Gupta I, Hoos A, Zhi E, Baron J, Piontek T, Lewis E, Jewell RC, Dettman EJ, Popat R, Esposti SD, Opalinska J, Richardson P, and Cohen AD
- Subjects
- Adolescent, Adult, Aged, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological adverse effects, Female, Humans, Male, Middle Aged, Multiple Myeloma mortality, Multiple Myeloma pathology, Time Factors, Treatment Outcome, Young Adult, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Multiple Myeloma drug therapy
- Abstract
Background: Belantamab mafodotin (GSK2857916), an immunoconjugate targeting B-cell maturation antigen, showed single-agent activity in the phase 1 DREAMM-1 study in heavily pre-treated patients with relapsed or refractory multiple myeloma. We further investigated the safety and activity of belantamab mafodotin in the DREAMM-2 study., Methods: DREAMM-2 is an open-label, two-arm, phase 2 study done at 58 multiple myeloma specialty centres in eight countries. Patients (aged ≥18 years) with relapsed or refractory multiple myeloma with disease progression after three or more lines of therapy and who were refractory to immunomodulatory drugs and proteasome inhibitors, and refractory or intolerant (or both) to an anti-CD38 monoclonal antibody with an Eastern Cooperative Oncology Group performance status of 0-2 were recruited, centrally randomly assigned (1:1) with permuted blocks (block size 4), and stratified by previous lines of therapy (≤4 vs >4) and cytogenetic features to receive 2·5 mg/kg or 3·4 mg/kg belantamab mafodotin via intravenous infusion every 3 weeks on day 1 of each cycle until disease progression or unacceptable toxicity. The intention-to-treat population comprised all randomised patients, regardless of treatment administration. The safety population comprised all patients who received at least one dose of belantamab mafodotin. The primary outcome was the proportion of randomly assigned patients in the intention-to-treat population who achieved an overall response, as assessed by an independent review committee. This study is registered with ClinicalTrials.gov, NCT03525678, and is ongoing., Findings: Between June 18, 2018, and Jan 2, 2019, 293 patients were screened and 196 were included in the intention-to-treat population (97 in the 2·5 mg/kg cohort and 99 in the 3·4 mg/kg cohort). As of June 21, 2019 (the primary analysis data cutoff date), 30 (31%; 97·5% CI 20·8-42·6) of 97 patients in the 2·5 mg/kg cohort and 34 (34%; 23·9-46·0) of 99 patients in the 3·4 mg/kg cohort achieved an overall response. The most common grade 3-4 adverse events in the safety population were keratopathy (in 26 [27%] of 95 patients in the 2·5 mg/kg cohort and 21 [21%] of 99 patients in the 3·4 mg/kg cohort), thrombocytopenia (19 [20%] and 33 [33%]), and anaemia (19 [20%] and 25 [25%]); 38 (40%) of 95 patients in the 2·5 mg/kg cohort and 47 (47%) of 99 in the 3·4 mg/kg cohort reported serious adverse events. Two deaths were potentially treatment related (one case of sepsis in the 2·5 mg/kg cohort and one case of haemophagocytic lymphohistiocytosis in the 3·4 mg/kg cohort)., Interpretation: Single-agent belantamab mafodotin shows anti-myeloma activity with a manageable safety profile in patients with relapsed or refractory multiple myeloma., Funding: GlaxoSmithKline., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
199. Daratumumab is effective in the relapsed or refractory systemic light-chain amyloidosis but associated with high infection burden in a frail real-life population.
- Author
-
Van de Wyngaert Z, Carpentier B, Pascal L, Lionne-Huyghe P, Leduc I, Srour M, Vasseur M, Demarquette H, Terriou L, Herbaux C, Manier S, Bossard JB, Barbieux S, Chauvet P, Willaume A, Nudel M, Bories C, Gibier JB, Facon T, and Boyle EM
- Subjects
- Aged, Cost of Illness, Female, Humans, Male, Middle Aged, Retrospective Studies, Antibodies, Monoclonal administration & dosage, Immunoglobulin Light-chain Amyloidosis drug therapy, Infections drug therapy
- Published
- 2020
- Full Text
- View/download PDF
200. A simplified frailty scale predicts outcomes in transplant-ineligible patients with newly diagnosed multiple myeloma treated in the FIRST (MM-020) trial.
- Author
-
Facon T, Dimopoulos MA, Meuleman N, Belch A, Mohty M, Chen WM, Kim K, Zamagni E, Rodriguez-Otero P, Renwick W, Rose C, Tempescul A, Boyle E, Manier S, Attal M, Moreau P, Macro M, Leleu X, Lorraine Chretien M, Ludwig H, Guo S, Sturniolo M, Tinel A, Silvia Monzini M, Costa B, Houck V, Hulin C, and Yves Mary J
- Subjects
- Adult, Aged, Aged, 80 and over, Dexamethasone administration & dosage, Dexamethasone adverse effects, Female, Frail Elderly, Humans, Lenalidomide administration & dosage, Lenalidomide adverse effects, Male, Melphalan administration & dosage, Melphalan adverse effects, Middle Aged, Prednisone administration & dosage, Prednisone adverse effects, Thalidomide administration & dosage, Thalidomide adverse effects, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Frailty, Multiple Myeloma drug therapy, Multiple Myeloma mortality
- Abstract
Patients with multiple myeloma are generally older and vary in fitness levels, which may influence the clinical benefit of treatment. Patients from the large, phase 3 FIRST trial in newly diagnosed multiple myeloma (NDMM) were retrospectively investigated to determine outcomes based on frailty using scores for age, Charlson Comorbidity Index (CCI), and Eastern Cooperative Oncology Group performance status (ECOG PS), instead of the EQ-5D quality-of-life questionnaire, as previously reported. ECOG PS (n = 1618) was investigated in frailty groups: frail (49%) and nonfrail (51%). Frail patients experienced worse progression-free and overall survival vs nonfrail patients. Prognostic assessment was improved when combining frailty and International Staging System stage (I/II vs III). Frail patients had a higher risk of developing grade 3/4 treatment-emergent adverse events. Treatment effects observed in the FIRST trial were confirmed per frailty group and per frailty and ISS group. The use of this ECOG PS-containing frailty scale as a predictive measure of clinical outcomes in patients with transplant-ineligible NDMM is supported by data from the FIRST trial. This score, based on age, CCI, and ECOG PS, can be easily replicated and may help design future myeloma studies in frail or nonfrail elderly patients.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.