1
|
Paiva B, Shi Q, Puig N, Cedena MT, Orfao A, Durie BGM, Munshi NC, San-Miguel J. Opportunities and challenges for MRD assessment in the clinical management of multiple myeloma. Nat Rev Clin Oncol 2025; 22:424-438. [PMID: 40195455 DOI: 10.1038/s41571-025-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
Measurable residual disease (MRD) assessment is, from the methodological point of view, ready for prime time in multiple myeloma (MM). Abundant evidence underscores the value of MRD status determined using highly sensitive next-generation flow cytometry and next-generation sequencing tests in evaluating response to treatment and, therefore, prognosis in patients with this disease. MRD response assessment and monitoring might present a range of opportunities for individualized patient management. Moreover, the considerable amounts of high-quality and standardized MRD data generated in clinical trials have led to the acceptance of MRD negativity as an early end point for accelerated regulatory approval of treatments for MM. The data leave no doubt that the efficacy of new regimens in inducing deeper and durable MRD-negative responses is connected with prolonged survival. Yet, several evidential, technical and practical challenges continue to limit the implementation of MRD-guided treatment strategies in routine practice, and the use of MRD as a surrogate end point remains controversial to some. In this Review, we draw on past and present research to propose opportunities for overcoming some of these challenges, and to accelerate the use of MRD assessment for improved clinical management of patients with MM.
Collapse
Affiliation(s)
- Bruno Paiva
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain.
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Noemi Puig
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00233, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Maria-Teresa Cedena
- Instituto de Investigación imas12, CIBER-ONC number CB16/12/00369, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service, Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00400, University of Salamanca, Salamanca, Spain
| | - Brian G M Durie
- Division of Hematology/Oncology, Cedars-Sinai Outpatient Cancer Center, Los Angeles, CA, USA
| | - Nikhil C Munshi
- Basic and Correlative Science, Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, VA Boston Healthcare System, Boston, MA, USA
| | - Jesús San-Miguel
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Zhang M, Cai T, Jin S, Huang X, Gao Y, Chen X, Ouyang W, Tao Y, Liu Y, Wang Y, Meng H, Li J, Lin X, Shi K, Wang J, Ding X, Mi JQ, Li B. Prognostic value of [ 18F]fluorodeoxyglucose-PET/MRI(CT) novel interpretation criteria (IMPeTUs) in multiple myeloma. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07219-w. [PMID: 40175849 DOI: 10.1007/s00259-025-07219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/13/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE [18F]fluorodeoxyglucose (FDG)-PET is a powerful tool to evaluate prognosis in multiple myeloma (MM). The development of systematic and reproducible standard interpretation criteria is crucial for the effective application of FDG-PET in MM. A new set of criteria-Italian Myeloma criteria for PET Use (IMPeTUs)-has standardized PET evaluation in MM. However, the prognostic value of IMPeTUs score remains unknown. METHODS A total of 58 patients with newly diagnosed multiple myeloma (NDMM) who underwent both [18F]FDG-PET/MRI and PET/CT examinations at diagnosis were enrolled (ChiCTR1900022597). All patients completed a 42-month follow-up. The prognostic value of the PET/MRI (or PET/CT) IMPeTUs score in predicting progression-free (PFS) and overall (OS) survival was compared with that of individual laboratory parameters and the maximum standardized uptake value (SUVmax). Kaplan-Meier method was used for survival analysis. Univariate and multivariate analyses of prognostic factors were conducted using Cox regression. RESULTS ROC curves demonstrated that the area under the curve for the PET/MRI IMPeTUs score was 0.760, exceeding that of the PET/CT IMPeTUs score (0.711), PET/CT BM SUVmax (0.649), PET/MRI BM SUVmax (0.575), bone marrow plasma cells (0.500), and β2-microglobulin (0.501). Univariate analysis and Kaplan-Meier analysis showed that a PET/MRI IMPeTUs score ≥ 13 and PET/CT IMPeTUs score ≥ 10 were significantly associated with worse PFS. Cox multivariate analysis showed that a PET/MRI IMPeTUs score ≥ 13 was an independent risk factor for PFS. CONCLUSION IMPeTUs standardized [18F]FDG-PET/MRI and PET/CT readings in MM. The IMPeTUs score is crucial for predicting MM prognosis, performing better than SUVmax and clinical indicators. CLINICAL TRIAL REGISTRATION ChiCTR1900022597, Registered on 18 April 2019.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Tangjia Cai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Shiwei Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinyun Huang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Yuting Gao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyue Chen
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Wanyan Ouyang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi Tao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuanfang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongping Meng
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Jian Li
- Clinical Research Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Xiaozhu Lin
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
| | - Kuangyu Shi
- Department of Nuclear Medicine, University of Bern, Bern, Switzerland
| | - Jin Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyi Ding
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- Institute for Medical Imaging Technology, Shanghai, 200025, China.
| |
Collapse
|
3
|
Barnidge D, Sakrikar D, Kubicki T, Derman BA, Jakubowiak AJ, Lakos G. Distinguishing Daratumumab from Endogenous Monoclonal Proteins in Serum from Multiple Myeloma Patients Using an Automated Mass Spectrometry System. J Appl Lab Med 2025; 10:235-249. [PMID: 39699179 DOI: 10.1093/jalm/jfae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Therapeutic monoclonal antibodies (t-mAbs) may interfere with electrophoresis-based methods used to monitor multiple myeloma (MM), which can create inaccurate results. Matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry is an alternative to gels distinguishing between endogenous M-proteins and t-mAbs based on molecular mass. METHODS Serum samples (n = 109) from 34 MM patients receiving Dara-KRd were collected 14 or 28 days postdaratumumab administration. Samples were analyzed using the EXENT® Analyzer that combines automated immunopurification and MALDI-TOF MS for the isotyping and quantification of monoclonal immunoglobulins. RESULTS Daratumumab was identified in 103 out of 109 samples (94.5%). In all IgGλ (n = 8), IgAκ (n = 8), and IgAλ (n = 2) patients, the M-protein and daratumumab were detected. Of the IgGκ patients (n = 18), 5 patients had a total of 6 samples where the M-protein was detected but daratumumab was not. There was no difference in the detection rate of daratumumab between samples taken 14 and 28 days postadministration with the median daratumumab concentration being 0.95 and 0.54 g/L, respectively. A precision study was also performed on 25 replicates containing 1 g/L daratumumab in serum where a coefficient of variation of 4.2% was observed as determined by the EXENT Analyzer. CONCLUSIONS The Immunoglobulin Isotypes (GAM: IgG, IgA, and IgM) for the EXENT Analyzer detected and distinguished a daratumumab kappa light chain peak from an M-protein light chain peak in MM patient serum when resolved by the mass spectrometer.
Collapse
Affiliation(s)
- David Barnidge
- The Binding Site, part of Thermo Fisher Scientific, Rochester, MN, United States
| | - Dhananjay Sakrikar
- The Binding Site, part of Thermo Fisher Scientific, Rochester, MN, United States
| | - Tadeusz Kubicki
- Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Benjamin A Derman
- Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | | | - Gabriella Lakos
- The Binding Site, part of Thermo Fisher Scientific, Rochester, MN, United States
| |
Collapse
|
4
|
Costa LJ, Gay F, Landgren O, Mateos MV, Moreau P, Touzeau C, Ertel F, McFadden I, Najdi R, Weisel K. Evolution of frontline treatment for multiple myeloma: clinical investigation of quadruplets containing carfilzomib and anti-CD38 monoclonal antibodies. Ann Hematol 2025; 104:1329-1351. [PMID: 39774926 PMCID: PMC12031813 DOI: 10.1007/s00277-024-06143-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025]
Abstract
Although survival rates for patients with newly diagnosed multiple myeloma (NDMM) have improved over recent decades, multiple myeloma (MM) remains without a cure for most. There is increasing consensus that achievement of deep remissions, especially minimal residual disease negativity (MRD -), in frontline treatment is crucial and translates into improved survival. The standard of care (SOC) for NDMM consists at minimum of a triplet regimen of therapies, with or without an autologous stem cell transplant, or a doublet regimen for certain ineligible, particularly frail patients who may have specific limitations. Recently, anti-CD38 monoclonal antibodies (mAbs), such as daratumumab (Dara) or isatuximab (Isa), have been integrated into frontline SOC regimens. Seeking to further deepen and prolong responses, several clinical trials have commenced investigating the addition of anti-CD38 mAbs to carfilzomib, lenalidomide, and dexamethasone (KRd). These quadruplet regimens (Isa/Dara-KRd) are being evaluated in the context of evolving treatment considerations for the heterogeneous population of patients with NDMM. In clinical trials, the addition of Isa/Dara to KRd achieved high rates of deep responses and MRD - . Favorable outcomes were observed in patients with NDMM independent of age, transplant eligibility, and cytogenetic risk, while these treatments did not result in unexpected or emergent safety risks. The efficacy observed with intensified, yet well-tolerated therapy may offer further development of risk- and response-adapted therapy for individualized patient needs. This review summarizes the clinical outcomes of quadruplet-based therapy with Isa/Dara-KRd in NDMM.
Collapse
Affiliation(s)
- Luciano J Costa
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham Hospital, 1802 6Th Avenue South, Birmingham, AL, 35294, USA.
| | - Francesca Gay
- AOU Città Della Salute E Della Scienza Di Torino, University of Torino, Turin, Italy
| | - Ola Landgren
- Division of Myeloma, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | | | - Philippe Moreau
- The Hematology Clinic, University Hospital Hôtel-Dieu, Nantes, France
| | - Cyrille Touzeau
- The Hematology Clinic, University Hospital Hôtel-Dieu, Nantes, France
| | | | | | | | - Katja Weisel
- Department of Oncology, Hematology and BMT, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Bhutani M, Robinson M, Foureau D, Atrash S, Paul B, Guo F, Grayson JM, Ivanina-Foureau A, Pineda-Roman M, Varga C, Friend R, Ferreri CJ, Begic X, Norek S, Drennan T, Anderson MB, Symanowski JT, Voorhees PM, Usmani SZ. MRD-driven phase 2 study of daratumumab, carfilzomib, lenalidomide, and dexamethasone in newly diagnosed multiple myeloma. Blood Adv 2025; 9:507-519. [PMID: 39576965 PMCID: PMC11814523 DOI: 10.1182/bloodadvances.2024014417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
ABSTRACT In newly diagnosed multiple myeloma (NDMM), measurable residual disease (MRD) status is prognostically important, but its role in treatment decisions remains unclear. In a phase 2 trial, we assessed daratumumab, carfilzomib, lenalidomide, and dexamethasone (Dara-KRd) induction followed by a next-generation sequencing-based MRD-adapted strategy. The primary outcome was complete response (CR) and stringent CR (≥CR) after induction. Flow cytometry was used to profile T cells. Among 39 patients, 21 (54%) achieved ≥CR after induction (P = .375), with MRD-negative rates of 59% (10-5) and 41% (10-6). Patients who were MRD-negative (n = 24, group A) received lenalidomide maintenance, showing sustained MRD negativity in 14 of 18 (77.8%) for ≥12 cycles. MRD-positive transplant-eligible patients (n = 8, group B) underwent autologous stem cell transplantation, with 62.5% converting to MRD-negative at 10-5 (37.5% at 10-6) posttransplant. MRD-positive, transplant-ineligible patients (n = 4, group C) received KRd consolidation. Best MRD-negative rates improved to 77% (10-5) and 72% (10-6). No new safety concerns were identified for Dara-KRd. With a median follow-up of 30.1 months, 3, 2, and 1 patient(s) in groups A, B, and C, respectively, have progressed or died. We observed that Dara-KRd strongly activated memory T cells, which was associated with an MRD-negative state post induction. Although the primary outcome was not met, Dara-KRd induction in NDMM achieved high ≥CR and MRD-negative rates without new safety concerns. The post induction MRD-adapted strategy deepened responses in MRD-positive patients and maintained durable MRD control in MRD-negative patients. This trial was registered at www.clinicaltrials.gov as #NCT04113018.
Collapse
Affiliation(s)
- Manisha Bhutani
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Myra Robinson
- Department of Biostatistics and Data Sciences, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - David Foureau
- Department of Internal Medicine, Section of Hematology and Oncology, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Shebli Atrash
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Barry Paul
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Fei Guo
- Department of Internal Medicine, Section of Hematology and Oncology, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Jason M. Grayson
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Anna Ivanina-Foureau
- Department of Internal Medicine, Section of Hematology and Oncology, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Mauricio Pineda-Roman
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Cindy Varga
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Reed Friend
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Christopher J. Ferreri
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Xhevahire Begic
- Clinical Trials Office, Wake Forest Baptist Comprehensive Cancer Center, Atrium Health Levine Cancer, Charlotte, NC
| | - Sarah Norek
- Clinical Trials Office, Wake Forest Baptist Comprehensive Cancer Center, Atrium Health Levine Cancer, Charlotte, NC
| | - Tiffany Drennan
- Clinical Trials Office, Wake Forest Baptist Comprehensive Cancer Center, Atrium Health Levine Cancer, Charlotte, NC
| | - Michelle B. Anderson
- Department of Biostatistics and Data Sciences, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - James T. Symanowski
- Department of Biostatistics and Data Sciences, Atrium Health Levine Cancer Institute, Charlotte, NC
| | - Peter M. Voorhees
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest University School of Medicine, Charlotte, NC
| | - Saad Z. Usmani
- Myeloma Service, Division of Hematologic Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
6
|
Kakkilaya A, Trando A, Cliff ERS, Mian H, Al Hadidi S, Aziz M, Goodman AM, Jeong AR, Smith WL, Kelkar AH, Russler-Germain DA, Mehra N, Chakraborty R, Gertz MA, Mohyuddin GR. Evaluating early intervention in smoldering myeloma clinical trials: a systematic review. Oncologist 2025; 30:oyae219. [PMID: 39236068 PMCID: PMC11883161 DOI: 10.1093/oncolo/oyae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Smoldering multiple myeloma (SMM), an asymptomatic precursor of multiple myeloma (MM), carries a variable risk of progression to MM. There is little consensus on the efficacy or optimal timing of treatment in SMM. We systematically reviewed the landscape of all clinical trials in SMM. We compared the efficacy of treatment regimens studied in SMM to results from these regimens when used in newly diagnosed multiple myeloma (NDMM), to determine whether the data suggest deeper responses in SMM versus NDMM. METHODS All prospective interventional clinical trials for SMM, including published studies, meeting abstracts, and unpublished trials listed on ClinicalTrials.gov up to April 1, 2023, were identified. Trial-related variables were captured, including treatment strategy and efficacy results. Relevant clinical endpoints were defined as overall survival (OS) and quality of life. RESULTS Among 45 SMM trials identified, 38 (84.4%) assessed active myeloma drugs, while 7 (15.6%) studied bone-modifying agents alone. Of 18 randomized trials in SMM, only one (5.6%) had a primary endpoint of OS; the most common primary endpoint was progression-free survival (n = 7, 38.9%). Among 32 SMM trials with available results, 9 (28.1%) met their prespecified primary endpoint, of which 5 were single-arm studies. Six treatment regimens were tested in both SMM and NDMM; 5 regimens yielded a lower rate of very good partial response rate or better (≥VGPR) in SMM compared to the corresponding NDMM trial (32% vs 63%, 43% vs 53%, 40% vs 63%, 86% vs 89%, 92% vs 95%, and 94% vs 87%, respectively). CONCLUSION In this systematic review of all prospective interventional clinical trials in SMM, we found significant variability in trial design, including randomization status, primary endpoints, and types of intervention used. Despite the statistical limitations, comparison of treatment regimens revealed no compelling evidence that the treatment is more effective when introduced early in SMM compared to NDMM.
Collapse
Affiliation(s)
- Apoorva Kakkilaya
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Aaron Trando
- School of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Edward R Scheffer Cliff
- Program on Regulation, Therapeutics and Law, Division of Pharmacoepidemiology and Pharmacoeconomics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Hira Mian
- Division of Hematology, McMaster University, Hamilton, Canada
| | - Samer Al Hadidi
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Muhammad Aziz
- Division of Gastroenterology and Hepatology, University of Toledo, Toledo, OH, United States
| | - Aaron M Goodman
- Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, CA, United States
| | - Ah-Reum Jeong
- Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, CA, United States
| | - Wade L Smith
- Mulford Health Science Library, University of Toledo, Toledo, OH, United States
| | - Amar H Kelkar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - David A Russler-Germain
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, United States
| | - Nikita Mehra
- Department of Medical Oncology, Cancer Institute (WIA), Chennai, India
| | - Rajshekhar Chakraborty
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Ghulam Rehman Mohyuddin
- Division of Hematology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
7
|
Patel J, Degann S, Ahmad M, Lesky L. Delayed diagnosis of multiple myeloma in a young patient: a call for vigilance in diagnosis. BMJ Case Rep 2025; 18:e262158. [PMID: 39773967 DOI: 10.1136/bcr-2024-262158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Multiple myeloma is a rare haematologic malignancy, representing about 1-2% of all cancers and 17% of haematologic malignancies in the US, predominantly affecting older adults and more common in African Americans (AAs) and men. Light-chain multiple myeloma, a subtype accounting for 15% of multiple myeloma cases, often has a more aggressive clinical course. This case report discusses a rare case of an AA female in her early 30s, diagnosed with light-chain multiple myeloma following a pathological rib fracture. Initial symptoms were atypical, and diagnosis was delayed due to her young age and lack of common multiple myeloma signs, such as anaemia and hypercalcaemia. Treatment included induction chemotherapy and autologous stem cell transplant, leading to complete remission. This case underscores the need for heightened clinical suspicion and thorough investigation in young patients presenting with unexplained bone lesions, highlighting the diverse presentations and challenges in diagnosing multiple myeloma in younger populations.
Collapse
Affiliation(s)
- Jigar Patel
- Internal Medicine, The George Washington University, Washington, DC, USA
| | - Seta Degann
- Internal Medicine, The George Washington University, Washington, DC, USA
| | | | - Linda Lesky
- Internal Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
8
|
Portuguese AJ, Banerjee R, Chen G, Reddi S, Cowan AJ. Novel Treatment Options for Multiple Myeloma. JCO Oncol Pract 2025:OP2400752. [PMID: 39772633 DOI: 10.1200/op-24-00752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Multiple myeloma (MM), the second most common hematologic malignancy in the United States, is characterized by repeated cycles of remission and relapse, with increasing resistance to treatment after each line of therapy. Despite the virtually incurable nature of MM, recent therapeutic breakthroughs have fundamentally reshaped its treatment landscape. This review explores evolving care paradigms, spanning from newly diagnosed MM to relapsed or refractory disease. In the frontline setting, treatment strategies have shifted beyond their traditional emphasis on autologous stem-cell transplant eligibility to a broader categorization of patients on the basis of their suitability for quadruplet therapy. In the relapsed/refractory setting, novel immunotherapies, including chimeric antigen receptor T-cell (CAR-T) therapies and bispecific antibodies, have revolutionized treatment, offering new hope for patients with previously limited options. Precision medicine is playing a growing role in MM treatment, with venetoclax showing significant efficacy in patients with t(11;14) translocation, advancing targeted therapy for this subgroup. On the horizon, investigational CAR-T products and cereblon E3 ligase modulators, such as mezigdomide and iberdomide, may provide faster, more durable responses compared with current therapies. In addition, belantamab mafodotin, an antibody-drug conjugate withdrawn from the US market in 2022, is on the verge of reapproval after positive results from recent randomized trials. While these therapies offer significant potential, challenges remain in managing toxicity, ensuring treatment accessibility, and optimizing sequencing strategies. As the therapeutic arsenal expands, the need for personalized MM treatment plans that balance efficacy with quality of life becomes even more essential.
Collapse
Affiliation(s)
- Andrew J Portuguese
- Fred Hutchinson Cancer Center, Seattle, WA
- University of Washington, Seattle, WA
| | - Rahul Banerjee
- Fred Hutchinson Cancer Center, Seattle, WA
- University of Washington, Seattle, WA
| | | | | | - Andrew J Cowan
- Fred Hutchinson Cancer Center, Seattle, WA
- University of Washington, Seattle, WA
| |
Collapse
|
9
|
Banerjee R, Sexton R, Cowan AJ, Rosenberg AS, Ailawadhi S, Rajkumar SV, Kumar S, Dispenzieri A, Lonial S, Durie BGM, Richardson PG, Usmani SZ, Hoering A, Orlowski RZ. Dexamethasone dose intensity does not impact outcomes in newly diagnosed multiple myeloma: a secondary SWOG analysis. Blood 2025; 145:75-84. [PMID: 39321347 DOI: 10.1182/blood.2024025939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
ABSTRACT Dexamethasone is a key component of induction for newly diagnosed multiple myeloma (NDMM), despite common toxicities, including hyperglycemia and insomnia. In the randomized ECOG E4A03 trial, dexamethasone 40 mg once weekly was associated with lower mortality than higher doses. However, the performance of dexamethasone dose reductions below this threshold with regard to progression-free survival (PFS) and overall survival (OS) in NDMM has not been fully characterized. We conducted a secondary pooled analysis of the SWOG 0777 and SWOG 1211 studies of NDMM, which used lenalidomide and dexamethasone (Rd) alone, with or without bortezomib, and with or without elotuzumab. The planned dexamethasone intensity was 40 to 60 mg weekly in all arms. Patients were categorized into FD-DEX (full-dose dexamethasone maintained throughout induction) or LD-DEX (lowered-dose dexamethasone or discontinuation; only permitted for grade 3+ toxicities per both study protocols). Of the 541 evaluated patients, the LD-DEX group comprised 373 patients (69%). There were no differences in PFS or OS between the FD-DEX and LD-DEX groups, which were balanced in terms of age, stage, and performance status. Predictors of PFS and OS in the multivariate models were treatment arm, age ≥70 years, and thrombocytopenia. FD-DEX did not significantly improve either outcome. Our study suggests that dexamethasone dose reductions are common in multiple myeloma, even within clinical trials. Given the many toxicities and unclear benefits of dexamethasone in the era of modern treatment regimens, dexamethasone dose reduction during NDMM induction warrants further prospective studies. These trials were registered at www.clinicaltrials.gov as #NCT00644228 and NCT01668719.
Collapse
Affiliation(s)
- Rahul Banerjee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology/Oncology, University of Washington, Seattle, WA
| | | | - Andrew J Cowan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology/Oncology, University of Washington, Seattle, WA
| | - Aaron S Rosenberg
- Division of Hematology and Oncology, University of California Davis, Davis, CA
| | | | | | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA
| | | | - Paul G Richardson
- Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Saad Z Usmani
- Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
10
|
Liao Y, Zhang W, Liu Y, Zhu C, Zou Z. The role of ubiquitination in health and disease. MedComm (Beijing) 2024; 5:e736. [PMID: 39329019 PMCID: PMC11424685 DOI: 10.1002/mco2.736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Ubiquitination is an enzymatic process characterized by the covalent attachment of ubiquitin to target proteins, thereby modulating their degradation, transportation, and signal transduction. By precisely regulating protein quality and quantity, ubiquitination is essential for maintaining protein homeostasis, DNA repair, cell cycle regulation, and immune responses. Nevertheless, the diversity of ubiquitin enzymes and their extensive involvement in numerous biological processes contribute to the complexity and variety of diseases resulting from their dysregulation. The ubiquitination process relies on a sophisticated enzymatic system, ubiquitin domains, and ubiquitin receptors, which collectively impart versatility to the ubiquitination pathway. The widespread presence of ubiquitin highlights its potential to induce pathological conditions. Ubiquitinated proteins are predominantly degraded through the proteasomal system, which also plays a key role in regulating protein localization and transport, as well as involvement in inflammatory pathways. This review systematically delineates the roles of ubiquitination in maintaining protein homeostasis, DNA repair, genomic stability, cell cycle regulation, cellular proliferation, and immune and inflammatory responses. Furthermore, the mechanisms by which ubiquitination is implicated in various pathologies, alongside current modulators of ubiquitination are discussed. Enhancing our comprehension of ubiquitination aims to provide novel insights into diseases involving ubiquitination and to propose innovative therapeutic strategies for clinical conditions.
Collapse
Affiliation(s)
- Yan Liao
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Wangzheqi Zhang
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Yang Liu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Chenglong Zhu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Zui Zou
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| |
Collapse
|
11
|
Ortiz de Landazuri I, Oliver-Caldés A, Español-Rego M, Agulló C, Contreras MT, Zabaleta A, Puig N, Cabañas V, González-Calle V, Zugasti I, Inogés S, Rodríguez Otero P, Martin-Antonio B, Reguera JL, López-Diaz de Cerio A, Aróstegui JI, Uribe-Herranz M, Benítez-Ribas D, Rodríguez-Lobato LG, González EA, Tovar N, Charry P, Navarro S, Rosiñol L, Tréboles K, Mora G, Yagüe J, Moraleda JM, Urbano-Ispizua Á, Mateos MV, Pascal M, Paiva B, Juan M, Fernández de Larrea C. Serum mass spectrometry for treatment monitoring in patients with multiple myeloma receiving ARI0002h CAR T-cells. Br J Haematol 2024; 205:1346-1355. [PMID: 38894496 DOI: 10.1111/bjh.19589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapies have increased the patients with relapsed/refractory multiple myeloma (RRMM) in whom standard electrophoretic techniques fail to detect the M-protein. Quantitative immunoprecipitation mass spectrometry (QIP-MS) can accurately measure serum M-protein with high sensitivity, and identify interferences caused by therapeutic monoclonal antibodies. Here, we investigate the outcome of QIP-MS in 33 patients treated with the academic BCMA-directed CAR T-cell ARI0002h (Cesnicabtagene Autoleucel). QIP-MS offered more detailed insights than serum immunofixation (sIFE), identifying glycosylated M-proteins and minor additional peaks. Moreover, the potential interferences owing to daratumumab or tocilizumab treatments were successfully detected. When analysing different assay platforms during patient's monitoring after ARI0002h administration, we observed that QIP-MS showed a high global concordance (78.8%) with sIFE, whereas it was only moderate (55.6%) with bone marrow (BM)-based next-generation flow cytometry (NGF). Furthermore, QIP-MS consistently demonstrated the lowest negativity rate across the different timepoints (27.3% vs. 60.0% in months 1 and 12, respectively). Patients with QIP-MS(+)/BM-based NGF(-) showed a non-significant shorter median progression free survival than those with QIP-MS(-)/BM-based NGF(-). In summary, we show the first experience to our knowledge demonstrating that QIP-MS could be particularly useful as a non-invasive technique when evaluating response after CAR T-cell treatment in MM.
Collapse
Affiliation(s)
- Iñaki Ortiz de Landazuri
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Aina Oliver-Caldés
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Hospital Universitari Son Espases, Palma de Mallorca, Spain
| | - Marta Español-Rego
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Cristina Agulló
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - María Teresa Contreras
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Aintzane Zabaleta
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Noemí Puig
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Valentín Cabañas
- Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Pascual Parrilla), Universidad de Murcia, Murcia, Spain
| | - Verónica González-Calle
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Inés Zugasti
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Susana Inogés
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Paula Rodríguez Otero
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | | | - Juan Luis Reguera
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC/CIBERONC), Universidad de Sevilla, Sevilla, Spain
| | - Ascensión López-Diaz de Cerio
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Juan Ignacio Aróstegui
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mireia Uribe-Herranz
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Daniel Benítez-Ribas
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Luis Gerardo Rodríguez-Lobato
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Europa Azucena González
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Natalia Tovar
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Paola Charry
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergio Navarro
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Laura Rosiñol
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Karen Tréboles
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Génesis Mora
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Yagüe
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - José María Moraleda
- Hospital Clínico Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria (IMIB-Pascual Parrilla), Universidad de Murcia, Murcia, Spain
| | - Álvaro Urbano-Ispizua
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - María Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Instituto Universitario de Biología Molecular y Celular del Cáncer (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Mariona Pascal
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Manel Juan
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Carlos Fernández de Larrea
- Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Kazandjian D, Diamond B, Papadimitriou M, Hill E, Sklavenitis-Pistofidis R, Ziccheddu B, Blaney P, Chojnacka M, Durante M, Maclachlan K, Young R, Usmani S, Davies F, Getz G, Ghobrial I, Korde N, Morgan G, Maura F, Landgren O. Genomic Profiling to Contextualize the Results of Intervention for Smoldering Multiple Myeloma. Clin Cancer Res 2024; 30:4482-4490. [PMID: 38652812 PMCID: PMC11444893 DOI: 10.1158/1078-0432.ccr-24-0210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/15/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Early intervention for high-risk smoldering multiple myeloma (HR-SMM) achieves deep and prolonged responses. It is unclear if beneficial outcomes are due to the treatment of less complex, susceptible disease or inaccuracy in clinical definition of cases entered. EXPERIMENTAL DESIGN In this study, we interrogated whole-genome and whole-exome sequencing for 54 patients across two HR-SMM interventional studies (NCT01572480 and NCT02279394). RESULTS We reveal that the genomic landscape of treated HR-SMM is generally simple as compared with newly diagnosed multiple myeloma counterparts with less inactivation of tumor suppressor genes, RAS pathway mutations, MYC disruption, and APOBEC contribution. The absence of these events parallels that of indolent precursor conditions, possibly explaining overall excellent outcomes. However, some patients harboring genomic complexity fail to sustain response and experience resistant, progressive disease. Overall, clinical risk scores do not effectively discriminate between genomically indolent and aggressive disease. CONCLUSIONS Genomic profiling can contextualize the advantage of early intervention in SMM and guide personalization of therapy. See related commentary by Weinhold and Rasche, p. 4263.
Collapse
Affiliation(s)
- Dickran Kazandjian
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Benjamin Diamond
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Marios Papadimitriou
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Elizabeth Hill
- Myeloma Program, Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Bachisio Ziccheddu
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Patrick Blaney
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY
| | - Monika Chojnacka
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Michael Durante
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ryan Young
- Myeloma Program, Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Saad Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Faith Davies
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Irene Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gareth Morgan
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY
| | - Francesco Maura
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Ola Landgren
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
13
|
Paul B, Anwer F, Raza S, Mammadzadeh A, Khasawneh B, Shatnawi S, McGuirk J, Ahmed N, Mahmoudjafari Z, Mushtaq M, Abdallah AO, Atrash S. Comparative Meta-Analysis of Triplet vs. Quadruplet Induction Regimens in Newly Diagnosed, Treatment Naïve, Multiple Myeloma. Cancers (Basel) 2024; 16:2938. [PMID: 39272795 PMCID: PMC11394295 DOI: 10.3390/cancers16172938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
The use of 4-drug induction regimens for treatment naïve newly diagnosed multiple myeloma (NDMM) is associated with improved depth of response and progression-free survival (PFS). However, head-to-head trials of 4-drug combinations are lacking, and instead, these regimens are typically compared to 3-drug backbones; limiting the ability to discern whether any additional benefit (or toxicity) is simply additive or represents a synergy (or interaction). We conducted a meta-analysis of phase 2 and phase 3 clinical trials that randomized treatment naïve NDMM patients to either a 4-drug or 3-drug induction regimen. We included 11 trials which represented 6509 unique patients. PFS for all trials in the meta-analysis was 54 months with a 4-drug induction and 8.9 months with a 3-drug induction (HR: 0.49; 95% CI: 0.45; 0.54), but there was no benefit to using a 4-drug induction that did not include an anti-CD38 antibody (PFS 4-drug 8.1 months, PFS 3-drug 8.0 months; HR 0.95; 95% CI 0.86; 1.06). Adverse events were more frequent with the quadruplet regimens but were predominately mild. High-grade (≥3) adverse events (AEs) that were more common with 4-drug regimens were infections (RR: 1.34; 95% CI 1.17; 1.54) and thrombocytopenia (RR: 1.39; 95% CI 1.12; 1.74). This study suggests that 4-drug induction regimens which include an anti-CD38 antibody improve efficacy although with additional toxicity in NDMM patients.
Collapse
Affiliation(s)
- Barry Paul
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health Wake Forest University School of Medicine, Charlotte, NC 28204, USA
| | - Faiz Anwer
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Shahzad Raza
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Aytaj Mammadzadeh
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Division of Hematology and Oncology, Mayo Clinic, Rochester, MN 44905, USA
| | - Bayan Khasawneh
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Department of Internal Medicine, Jordan University of Science & Technology, Irbid 22110, Jordan
| | - Sara Shatnawi
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Department of Internal Medicine, Jordan University of Science & Technology, Irbid 22110, Jordan
| | - Joseph McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS 66205, USA
| | - Nausheen Ahmed
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS 66205, USA
| | - Zahra Mahmoudjafari
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS 66205, USA
| | - Muhammad Mushtaq
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS 66205, USA
| | - Al-Ola Abdallah
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS 66205, USA
| | - Shebli Atrash
- US Myeloma Innovations Research Collaborative (USMIRC), Westwood, KS 66205, USA
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health Wake Forest University School of Medicine, Charlotte, NC 28204, USA
| |
Collapse
|
14
|
Cirrincione AM, Poos AM, Ziccheddu B, Kaddoura M, Bärtsch MA, Maclachlan K, Chojnacka M, Diamond B, John L, Reichert P, Huhn S, Blaney P, Gagler D, Rippe K, Zhang Y, Dogan A, Lesokhin AM, Davies F, Goldschmidt H, Fenk R, Weisel KC, Mai EK, Korde N, Morgan GJ, Usmani S, Landgren O, Raab MS, Weinhold N, Maura F. The biological and clinical impact of deletions before and after large chromosomal gains in multiple myeloma. Blood 2024; 144:771-783. [PMID: 38728430 PMCID: PMC11375460 DOI: 10.1182/blood.2024024299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/16/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
ABSTRACT Acquisition of a hyperdiploid (HY) karyotype or immunoglobulin heavy chain (IgH) translocations are considered key initiating events in multiple myeloma (MM). To explore if other genomic events can precede these events, we analyzed whole-genome sequencing data from 1173 MM samples. By integrating molecular time and structural variants within early chromosomal duplications, we indeed identified pregain deletions in 9.4% of patients with an HY karyotype without IgH translocations, challenging acquisition of an HY karyotype as the earliest somatic event. Remarkably, these deletions affected tumor suppressor genes (TSGs) and/or oncogenes in 2.4% of patients with an HY karyotype without IgH translocations, supporting their role in MM pathogenesis. Furthermore, our study points to postgain deletions as novel driver mechanisms in MM. Using multiomics approaches to investigate their biologic impact, we found associations with poor clinical outcome in newly diagnosed patients and profound effects on both the oncogene and TSG activity despite the diploid gene status. Overall, this study provides novel insights into the temporal dynamics of genomic alterations in MM.
Collapse
Affiliation(s)
| | - Alexandra M. Poos
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital and Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Bachisio Ziccheddu
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Marcella Kaddoura
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Marc-Andrea Bärtsch
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital and Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Monika Chojnacka
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Benjamin Diamond
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Lukas John
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital and Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Reichert
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Stefanie Huhn
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Patrick Blaney
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Dylan Gagler
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | - Yanming Zhang
- Cytogenetics Laboratory, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Dogan
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander M. Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Faith Davies
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Roland Fenk
- Department of Hematology, Oncology and Clinical Immunology, University-Hospital Duesseldorf, Duesseldorf, Germany
| | - Katja C. Weisel
- Department of Oncology, Hematology, and Blood and Marrow Transplant, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elias K. Mai
- Department of Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gareth J. Morgan
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Saad Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Marc S. Raab
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital and Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Niels Weinhold
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital and Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Francesco Maura
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
15
|
Akhmedov M, Zeynalova P, Fedenko A. Multiple myeloma and infections in the era of novel treatment modalities. Leuk Res 2024; 143:107544. [PMID: 38963989 DOI: 10.1016/j.leukres.2024.107544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/14/2024] [Accepted: 06/17/2024] [Indexed: 07/06/2024]
Abstract
Infections are major cause of morbidity and mortality in patients with multiple myeloma. Current treatment landscape of newly-diagnosed multiple myeloma includes different classes of drugs, such as proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies, all of which are characterized by specific risk and pattern of infectious complications. Additionally, autologous and allogeneic hematopoietic cell transplantation, widely used in the treatment of multiple myeloma, are complex procedures, carrying a significant risk of complications, and mainly infections. Finally, novel treatment modalities such as bispecific T-cell engagers and chimeric antigen receptor T-lymphocytes have been changing the paradigm of myeloma treatment in relapsed-refractory setting. These agents due to unique mechanism of action carry distinct pattern of infectious complications. In this review, an attempt has been made to summarize the incidence, risk factors, and patterns of infections during different stages of myeloma treatment including novel treatment modalities, and to provide evidence underlying the current concept of infectious disease prophylaxis in this category of patients.
Collapse
Affiliation(s)
- Mobil Akhmedov
- Department of High-dose Chemotherapy and Bone Marrow Transplantation, P.A. Herzen Moscow Oncology Research Institute, branch of the National Medical Radiology Research Center, Russian Federation; Department of Oncology and Oncosurgery, Russian University of Medicine, Russian Federation.
| | - Pervin Zeynalova
- Department of Oncology, Sechenov University, Russian Federation; Department of Oncology, Lapino Clinical Hospital, Russian Federation
| | - Alexander Fedenko
- Department of High-dose Chemotherapy and Bone Marrow Transplantation, P.A. Herzen Moscow Oncology Research Institute, branch of the National Medical Radiology Research Center, Russian Federation
| |
Collapse
|
16
|
Ziccheddu B, Giannotta C, D'Agostino M, Bertuglia G, Saraci E, Oliva S, Genuardi E, Papadimitriou M, Diamond B, Corradini P, Coffey D, Landgren O, Bolli N, Bruno B, Boccadoro M, Massaia M, Maura F, Larocca A. Genomic and immune determinants of resistance to daratumumab-based therapy in relapsed refractory multiple myeloma. Blood Cancer J 2024; 14:117. [PMID: 39030183 PMCID: PMC11271515 DOI: 10.1038/s41408-024-01096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Targeted immunotherapy combinations, including the anti-CD38 monoclonal antibody (MoAb) daratumumab, have shown promising results in patients with relapsed/refractory multiple myeloma (RRMM), leading to a considerable increase in progression-free survival. However, a large fraction of patients inevitably relapse. To understand this, we investigated 32 relapsed MM patients treated with daratumumab, lenalidomide, and dexamethasone (Dara-Rd; NCT03848676). We conducted an integrated analysis using whole-genome sequencing (WGS) and flow cytometry in patients with RRMM. WGS before and after treatment pinpointed genomic drivers associated with early progression, including RPL5 loss, APOBEC mutagenesis, and gain of function structural variants involving MYC and chromothripsis. Flow cytometry on 202 blood samples, collected every 3 months until progression for 31 patients, revealed distinct immune changes significantly impacting clinical outcomes. Progressing patients exhibited significant depletion of CD38-positive NK cells, persistence of T-cell exhaustion, and reduced depletion of regulatory T cells over time. These findings underscore the influence of immune composition and daratumumab-induced immune changes in promoting MM resistance. Integrating genomics and flow cytometry unveiled associations between adverse genomic features and immune patterns. Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd in patients with RRMM.
Collapse
Affiliation(s)
- Bachisio Ziccheddu
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Claudia Giannotta
- Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, Università di Torino, Torino, Italy
| | - Mattia D'Agostino
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Giuseppe Bertuglia
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Elona Saraci
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Stefania Oliva
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Elisa Genuardi
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Marios Papadimitriou
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Benjamin Diamond
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Paolo Corradini
- Division of Hematology and Bone Marrow Transplant, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - David Coffey
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Ola Landgren
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Benedetto Bruno
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | | | - Massimo Massaia
- Laboratory of Blood Tumor Immunology, Molecular Biotechnology Center "Guido Tarone", Department of Molecular Biotechnology and Health Sciences, Università di Torino, Torino, Italy
- SC Ematologia, AO S. Croce e Carle, Cuneo, Italy
| | - Francesco Maura
- Myeloma Division, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| | - Alessandra Larocca
- Division of Hematology, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| |
Collapse
|
17
|
Alhasan BA, Morozov AV, Guzhova IV, Margulis BA. The ubiquitin-proteasome system in the regulation of tumor dormancy and recurrence. Biochim Biophys Acta Rev Cancer 2024; 1879:189119. [PMID: 38761982 DOI: 10.1016/j.bbcan.2024.189119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/20/2024]
Abstract
Tumor recurrence is a mechanism triggered in sparse populations of cancer cells that usually remain in a quiescent state after strict stress and/or therapeutic factors, which is affected by a variety of autocrine and microenvironmental cues. Despite thorough investigations, the biology of dormant and/or cancer stem cells is still not fully elucidated, as for the mechanisms of their reawakening, while only the major molecular patterns driving the relapse process have been identified to date. These molecular patterns profoundly interfere with the elements of cellular proteostasis systems that support the efficiency of the recurrence process. As a major proteostasis machinery, we review the role of the ubiquitin-proteasome system (UPS) in tumor cell dormancy and reawakening, devoting particular attention to the functions of its components, E3 ligases, deubiquitinating enzymes and proteasomes in cancer recurrence. We demonstrate how UPS components functionally or mechanistically interact with the pivotal proteins implicated in the recurrence program and reveal that modulators of the UPS hold promise to become an efficient adjuvant therapy for eradicating refractory tumor cells to impede tumor relapse.
Collapse
Affiliation(s)
- Bashar A Alhasan
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia.
| | - Alexey V Morozov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Street 32, 119991 Moscow, Russia.
| | - Irina V Guzhova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia.
| | - Boris A Margulis
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia.
| |
Collapse
|
18
|
Hultcrantz M, Hassoun H, Korde N, Maclachlan K, Mailankody S, Patel D, Shah U, Tan CR, Chung DJ, Lahoud O, Landau H, Scordo M, Shah GL, Giralt S, Pianko MJ, Burge M, Barnett K, Salcedo M, Caple J, Tran L, Blaslov J, Shekarkhand T, Hamid S, Nemikovski D, Derkach A, Arisa O, Peer CJ, Figg WD, Usmani SZ, Landgren O, Lesokhin AM. Colesevelam for Lenalidomide Associated Diarrhea in Patients with Multiple Myeloma. RESEARCH SQUARE 2024:rs.3.rs-4406606. [PMID: 38883739 PMCID: PMC11177961 DOI: 10.21203/rs.3.rs-4406606/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Lenalidomide maintenance is associated with a significantly improved progression-free in patients with newly diagnosed multiple myeloma. Maintenance with lenalidomide is generally well tolerated; however, lenalidomide associated diarrhea is a common side effect and bile acid malabsorption has been suggested as an underlying mechanism. We conducted a single arm phase 2 trial of colesevelam, a bile acid binder, for lenalidomide-associated diarrhea in multiple myeloma. Patients were treated with colesevelam daily starting at 1250 mg (2 tablets 625 mg) for 12 weeks. The trial included 25 patients, 1 patient with grade 3 diarrhea, 14 with grade 2, and 10 with grade 1 diarrhea. All patients were on treatment with single agent lenalidomide maintenance and no patient progressed during the trial. Colesevelam treatment was highly effective for treatment of lenalidomide-associated diarrhea; 22 (88%) of the 25 patients responded where 17 patients (68%) had complete resolution of diarrhea, and 5 patients (20%) had improvement by 1 grade of diarrhea. The responses to colesevelam were seen within the first two weeks of treatment. These findings support the conclusion that lenalidomide-associated diarrhea is driven by bile acid malabsorption. Five patients reported mild gastrointestinal side effects including constipation. Importantly, the pharmacokinetics of lenalidomide were not affected by concomitant colesevelam treatment. The stool microbiome composition was not significantly different before and after colesevelam treatment. Patients reported improved diarrhea, fewer gastrointestinal symptoms, and less interference with their daily life after starting colesevelam. In summary, colesevelam was safe and highly effective for treatment of lenalidomide-associated diarrhea in multiple myeloma and does not reduce the clinical effect of lenalidomide.
Collapse
Affiliation(s)
- Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dhwani Patel
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Urvi Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carlyn Rose Tan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David J. Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Oscar Lahoud
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Heather Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sergio Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Matthew J Pianko
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Miranda Burge
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kelly Barnett
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Meghan Salcedo
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Julia Caple
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Linh Tran
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jenna Blaslov
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tala Shekarkhand
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Selena Hamid
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David Nemikovski
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andriy Derkach
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Oluwatobi Arisa
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Cody J Peer
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Saad Z Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Alexander M Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
19
|
Schmidt TM. Extended DaraKRd: Are we enhancing outcomes by prolonging treatment? Blood Cancer J 2024; 14:93. [PMID: 38834650 PMCID: PMC11150380 DOI: 10.1038/s41408-024-01070-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024] Open
|
20
|
Meseha M, Hoffman J, Kazandjian D, Landgren O, Diamond B. Minimal Residual Disease-Adapted Therapy in Multiple Myeloma: Current Evidence and Opinions. Curr Oncol Rep 2024; 26:679-690. [PMID: 38676789 PMCID: PMC11169024 DOI: 10.1007/s11912-024-01537-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2024] [Indexed: 04/29/2024]
Abstract
PURPOSE OF REVIEW Multiple myeloma (MM) is a biologically heterogeneous malignancy with relatively uniform treatment paradigms. This review aims to assess the growing role of Minimal Residual Disease (MRD) assessment in facilitating response-adapted therapeutic decision making to individualize therapy in MM. RECENT FINDINGS MRD has been repeatedly demonstrated to provide strong prognostic information, superseding traditional IMWG response criteria. The use of MRD to modulate therapy remains controversial. Here, we review the existing landscape of MRD-adapted trial designs in both induction/consolidation and maintenance settings, including recent data from influential studies and retrospective analyses. We navigate existing data, leverage the increased resolution of longitudinal MRD assessments, and comment on trials in progress to explain our current utilization of MRD in the clinic. MRD transcends traditional response assessments by providing a window into disease-treatment interaction over time. As a strong patient-level surrogate, MRD has limited current use in individualizing treatment, but is poised to comprehensively shape treatment strategies at many key points in a patient's MM course.
Collapse
Affiliation(s)
- Mina Meseha
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, 1120 NW 14th Street, Clinical Research Building, Miami, FL, 33136, USA
| | - James Hoffman
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, 1120 NW 14th Street, Clinical Research Building, Miami, FL, 33136, USA
| | - Dickran Kazandjian
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, 1120 NW 14th Street, Clinical Research Building, Miami, FL, 33136, USA
| | - Ola Landgren
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, 1120 NW 14th Street, Clinical Research Building, Miami, FL, 33136, USA
| | - Benjamin Diamond
- Myeloma Institute, Sylvester Comprehensive Cancer Center, University of Miami, 1120 NW 14th Street, Clinical Research Building, Miami, FL, 33136, USA.
| |
Collapse
|
21
|
Callander NS. Another quadruplet therapy for multiple myeloma: the beginning of the end for autologous haematopoietic stem-cell transplantation? Lancet Haematol 2024; 11:e392-e393. [PMID: 38677301 DOI: 10.1016/s2352-3026(24)00097-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/29/2024]
Affiliation(s)
- Natalie Scott Callander
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, Wisconsin Institutes for Medical Research, Madison, WI, USA, 53705-2275.
| |
Collapse
|
22
|
O'Donnell E, Mo C, Yee AJ, Nadeem O, Laubach J, Rosenblatt J, Munshi N, Midha S, Cirstea D, Chrysafi P, Horick N, Richardson PG, Raje N. Isatuximab, carfilzomib, lenalidomide, and dexamethasone in patients with newly diagnosed, transplantation-eligible multiple myeloma (SKylaRk): a single-arm, phase 2 trial. Lancet Haematol 2024; 11:e415-e424. [PMID: 38677302 DOI: 10.1016/s2352-3026(24)00070-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND Isatuximab is a CD38 monoclonal antibody approved for relapsed or refractory multiple myeloma. We aimed to evaluate the addition of isatuximab to weekly carfilzomib (K), lenalidomide (R), and dexamethasone (d; Isa-KRd) in transplant-eligible patients with newly diagnosed multiple myeloma and stratified maintenance by cytogenetic risk. METHODS This single-arm phase 2 trial was done at three cancer centres (two hospitals and a cancer institute) in Boston (MA, USA). Eligible patients were aged at least 18 years and had transplant-eligible newly diagnosed multiple myeloma and an ECOG performance status of 2 or less. Patients received four 28-day cycles of Isa-KRd, including isatuximab 10 mg/kg intravenously weekly for 8 weeks, then every other week for 16 weeks, and every 4 weeks thereafter; carfilzomib 56 mg/m2 intravenously on days 1, 8, and 15 (20 mg/m2 for cycle 1 day 1); lenalidomide 25 mg orally on days 1-21; and dexamethasone 20 mg orally the day of and day after all doses of carfilzomib and isatuximab. Consolidation involved either upfront haematopoietic stem-cell transplantation (HSCT) with two additional cycles or deferred HSCT with four additional cycles of treatment. The primary endpoint was complete response after four cycles of treatment. Analyses were by intention-to-treat. All patients who received one dose of study drug were included in the safety analyses. This study was registered at ClinicalTrials.gov, NCT04430894, and has completed enrolment. FINDINGS Between July 31, 2020 and Jan 31, 2022, 50 patients were enrolled. Median age was 59 years (range 40-70), 54% (27 of 50 patients) were male, and 44 (88%) were White. 46% (23 of 50) of patients had high-risk cytogenetics. Median follow-up was 26 months (IQR 20·7-30·1). 32% (16 of 50 patients) achieved a complete response after four cycles. The overall response rate (ORR) was 90% (45 patients) and 78% (39 patients) achieved a very good partial response (VGPR) or better. After completion of consolidation, 58% (29 patients) achieved a complete response; the ORR was 90% (45 patients) and 86% (43 patients) achieved a VGPR or better. The most common grade 3 or 4 side-effects (≥two patients) included neutropenia (13 [26%] of 50 patients), elevated alanine aminotransferase (six [12%] patients), fatigue (three [6%] patients), thrombocytopenia (three [6%] patients), acute kidney injury (two [4%] patients), anaemia (two [4%] patients), and febrile neutropenia (two [4%] patients). Grade 1-2 infusion-related reactions were seen in 20% (ten patients), with none grade 3. Grade 1-2 hypertension was seen in 14% (seven patients) with one grade 3 (one [2%] patient). There were two deaths assessed as unrelated to treatment. INTERPRETATION Although the study did not achieve the prespecified complete response threshold, Isa-KRd induced deep and durable responses in transplant-eligible patients with newly diagnosed multiple myeloma. The treatment proved safe and consistent with similar regimens in this setting. FUNDING Amgen, Sanofi, and Adaptive.
Collapse
Affiliation(s)
- Elizabeth O'Donnell
- Massachusetts General Hospital Cancer Center, Boston, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Clifton Mo
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Andrew J Yee
- Massachusetts General Hospital Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Omar Nadeem
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jacob Laubach
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Nikhil Munshi
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Shonali Midha
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Diana Cirstea
- Massachusetts General Hospital Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Nora Horick
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Paul G Richardson
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Noopur Raje
- Massachusetts General Hospital Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Derman BA, Cooperrider J, Rosenblatt J, Avigan DE, Rampurwala M, Barnidge D, Major A, Karrison T, Jiang K, Ramsland A, Kubicki T, Jakubowiak AJ. Final analysis of a phase II trial of daratumumab, carfilzomib, lenalidomide, and dexamethasone in newly diagnosed multiple myeloma without transplant. Blood Cancer J 2024; 14:87. [PMID: 38811560 PMCID: PMC11136961 DOI: 10.1038/s41408-024-01045-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 05/31/2024] Open
Abstract
We evaluated the efficacy and safety of 24 cycles of Dara in combination with carfilzomib (K), lenalidomide (R), and dexamethasone (d) without autologous stem cell transplant (ASCT) in newly diagnosed multiple myeloma (NDMM) irrespective of ASCT eligibility in a single-arm, phase II study. The primary endpoint was the rate of stringent complete response (sCR) and/or measurable residual disease (MRD) < 10-5 by next-generation sequencing (NGS) at the end of cycle 8 (C8). MRD was also assessed on peripheral blood samples using both the EXENT® system and liquid chromatography-mass spectrometry (LC-MS). Forty-two patients entered the treatment phase; forty were evaluable for the primary endpoint. The rate of sCR and/or MRD < 10-5 following C8 was 30/40 (75%), meeting the statistical threshold for efficacy. The 10-6 MRD negative rate improved with treatment beyond C8. Agreement between EXENT® and NGS was high and increased over time; agreement between LC-MS and NGS was lower. The estimated 3-year progression-free survival progression-free survival was 85%, and 3-year overall survival was 95%. Upper respiratory infections occurred in 67% (7% grade 3-4). There were no treatment-related deaths. Extended frontline Dara-KRd induced a high rate of sCR and/or MRD negativity; the rate and depth of MRD negativity improved beyond C8.
Collapse
Affiliation(s)
- Benjamin A Derman
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | | | | | | | | | - David Barnidge
- The Binding Site Group, part of Thermo Fisher, Rochester, MN, USA
| | | | - Theodore Karrison
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | - Ken Jiang
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | | | - Tadeusz Kubicki
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
24
|
Barbieri E, Martino EA, Rivolti E, Quaresima M, Vigna E, Neri A, Morabito F, Gentile M. Anti-CD38 monoclonal antibodies in multiple myeloma with gain/amplification of chromosome arm 1q: a review of the literature. Expert Opin Biol Ther 2024; 24:365-381. [PMID: 38757726 DOI: 10.1080/14712598.2024.2357382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
INTRODUCTION Gain/amplification of 1q (+1q) represents one of the most prevalent cytogenetic abnormalities (CAs) observed in multiple myeloma (MM). Historical studies predating the advent of anti-CD38 monoclonal antibodies (moAbs) implicated + 1q in poor prognoses, prompting its integration into novel staging systems. However, with the emergence of daratumumab and isatuximab, two pivotal anti-CD38 moAbs, the landscape of MM therapy has undergone a profound transformation. AREAS COVERED This review encompasses a comprehensive analysis of diverse study methodologies, including observational investigations, clinical trials, meta-analyses, and real-world database analyses. By synthesizing these data sources, we aim to provide an overview of the current understanding of + 1q in the context of anti-CD38 moAbs therapies. EXPERT OPINION Despite the paucity of available data, evidence suggests a potential mitigating effect of daratumumab on the adverse prognostic implications of + 1q. However, this benefit seems to diminish in patients harboring ≥ 4 copies or with concurrent high-risk CAs. On the other hand, isatuximab demonstrated promising outcomes in the relapsed-refractory setting for + 1q MM patients. Nevertheless, direct comparison between the two compounds is currently challenging. The current evidence firmly supports the integration of anti-CD38 moAb-based therapies as the standard of care for + 1q patients, pending further elucidation.
Collapse
Affiliation(s)
- Emiliano Barbieri
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Elena Rivolti
- Hematology Unit, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy
| | - Micol Quaresima
- Hematology Unit, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy
| | - Ernesto Vigna
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Antonino Neri
- Scientific Directorate, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Massimo Gentile
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
25
|
Coffey DG, Osman K, Aleman A, Bekri S, Kats S, Dhadwal A, Catamero D, Kim-Schulze S, Gnjatic S, Chari A, Parekh S, Jagannath S, Cho HJ. Phase 1 study combining elotuzumab with autologous stem cell transplant and lenalidomide for multiple myeloma. J Immunother Cancer 2024; 12:e008110. [PMID: 38609316 PMCID: PMC11029259 DOI: 10.1136/jitc-2023-008110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Autologous stem cell transplantation (ASCT) after induction therapy improves disease-free survival for patients with multiple myeloma (MM). While the goal of ASCT is to render a minimal disease state, it is also associated with eradication of immunosuppressive cells, and we hypothesize that early introduction of immunotherapy post-ASCT may provide a window of opportunity to boost treatment efficacy. METHODS We conducted a phase 1 clinical trial to investigate the application of autologous lymphocyte infusion and anti-SLAMF7 monoclonal antibody, elotuzumab, after ASCT in patients with newly diagnosed MM previously treated with induction therapy. In addition to CD34+ stem cells, peripheral blood mononuclear cells were harvested prior to transplant and infused on day 3 after stem cell infusion to accelerate immune reconstitution and provide autologous natural killer (NK) cells that are essential to the mechanism of elotuzumab. Elotuzumab was administered starting on day 4 and then every 28 days after until 1 year post-ASCT. Cycles 4-12 were administered with standard-of-care lenalidomide maintenance. RESULTS All subjects were evaluated for safety, and 13 of 15 subjects completed the treatment protocol. At 1 year post-ASCT, the disease status of enrolled subjects was as follows: five stringent complete responses, one complete response, six very good partial responses, one partial response, and two progressive diseases. The treatment plan was well tolerated, with most grade 3 and 4 AEs being expected hematologic toxicities associated with ASCT. Correlative analysis of the immune microenvironment demonstrated a trend toward reduced regulatory T cells during the first 3 months post-transplant followed by an increase in NK cells and monocytes in patients achieving a complete remission. CONCLUSIONS This phase 1 clinical trial demonstrates that early introduction of immunotherapy after ASCT is well tolerated and shows promising disease control in patients with MM, accompanied by favorable changes in the immune microenvironment. TRIAL REGISTRATION NUMBER NCT02655458.
Collapse
Affiliation(s)
- David G Coffey
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Keren Osman
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Adolfo Aleman
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Selma Bekri
- Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Simone Kats
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Amishi Dhadwal
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Donna Catamero
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Seunghee Kim-Schulze
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Sacha Gnjatic
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
- Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Samir Parekh
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Sundar Jagannath
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Hearn Jay Cho
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| |
Collapse
|
26
|
Nishida H, Suzuki R, Nakajima K, Hayashi M, Morimoto C, Yamada T. HDAC Inhibition Induces CD26 Expression on Multiple Myeloma Cells via the c-Myc/Sp1-mediated Promoter Activation. CANCER RESEARCH COMMUNICATIONS 2024; 4:349-364. [PMID: 38284882 PMCID: PMC10854391 DOI: 10.1158/2767-9764.crc-23-0215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/13/2023] [Accepted: 01/24/2024] [Indexed: 01/30/2024]
Abstract
CD26 is ubiquitously and intensely expressed in osteoclasts in patients with multiple myeloma, whereas its expression in plasma cells of patients with multiple myeloma is heterogeneous because of its cellular diversity, immune escape, and disease progression. Decreased expression levels of CD26 in myeloma cells constitute one of the mechanisms underlying resistance to humanized anti-CD26 mAb therapy in multiple myeloma. In the current study, we show that histone deacetylase inhibition (HDACi) with broad or class-specific inhibitors involves the induction of CD26 expression on CD26neg myeloma cells both transcriptionally and translationally. Furthermore, dipeptidyl peptidase Ⅳ (DPPⅣ) enzymatic activity was concomitantly enhanced in myeloma cells. Combined treatment with HDACi plus CD26mAb synergistically facilitated lysis of CD26neg myeloma cells not only by antibody-dependent cellular cytotoxicity but also by the direct effects of mAb. Of note, its combination readily augmented lysis of CD26neg cell populations, refractory to CD26mAb or HDACi alone. Chromatin immunoprecipitation assay revealed that HDACi increased acetylation of histone 3 lysine 27 at the CD26 promoter of myeloma cells. Moreover, in the absence of HDACi, c-Myc was attached to the CD26 promoter via Sp1 on the proximal G-C box of myeloma cells, whereas, in the presence of HDACi, c-Myc was detached from Sp1 with increased acetylation of c-Myc on the promoter, leading to activation of the CD26 promoter and initiation of transcription in myeloma cells. Collectively, these results confirm that HDACi plays crucial roles not only through its anti-myeloma activity but by sensitizing CD26neg myeloma cells to CD26mAb via c-Myc/Sp1-mediated CD26 induction, thereby augmenting its cytotoxicity. SIGNIFICANCE There is a desire to induce and sustain CD26 expression on multiple myeloma cells to elicit superior anti-myeloma response by humanized anti-CD26 mAb therapy. HDACi upregulates the expression levels of CD26 on myeloma cells via the increased acetylation of c-MycK323 on the CD26 promoter, leading to initiation of CD26 transcription, thereby synergistically augments the efficacy of CD26mAb against CD26neg myeloma cells.
Collapse
Grants
- 20K07682,16K07180 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and technology of Japan (C)
- 19H03519 Grant-in Aid for Scientific Research from the Ministry of Education, Culture, Sports and technology of Japan (B)
- 19K22542 Grant-in-Aid for Exploratory Research form the Ministry of Education, Culture Sports, Science and Technology of Japan
- 19H03519 Grant-in Aid for Scientific Research from the Ministry of Education, Culture, Sports and technology of Japan (B)
- 19K22542 Grant-in-Aid for Exploratory Research form the Ministry of Education, Culture Sports, Science and Technology of Japan
Collapse
Affiliation(s)
- Hiroko Nishida
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Division of Hematology, Department of Internal of Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Reiko Suzuki
- Department of Collaborative Research Resources, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kiyora Nakajima
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Mutsumi Hayashi
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Chikao Morimoto
- Department of Pathology, Faculty of Medicine, Saitama Medical University, Saitama, Japan
| | - Taketo Yamada
- Department of Pathology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Juntendo University, Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
27
|
Leypoldt LB, Tichy D, Besemer B, Hänel M, Raab MS, Mann C, Munder M, Reinhardt HC, Nogai A, Görner M, Ko YD, de Wit M, Salwender H, Scheid C, Graeven U, Peceny R, Staib P, Dieing A, Einsele H, Jauch A, Hundemer M, Zago M, Požek E, Benner A, Bokemeyer C, Goldschmidt H, Weisel KC. Isatuximab, Carfilzomib, Lenalidomide, and Dexamethasone for the Treatment of High-Risk Newly Diagnosed Multiple Myeloma. J Clin Oncol 2024; 42:26-37. [PMID: 37753960 PMCID: PMC10730063 DOI: 10.1200/jco.23.01696] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
PURPOSE The GMMG-CONCEPT trial investigated isatuximab, carfilzomib, lenalidomide, and dexamethasone (Isa-KRd) in transplant-eligible (TE) and transplant-noneligible (TNE) patients with newly diagnosed multiple myeloma (NDMM) with exclusively high-risk disease for whom prospective trials are limited, aiming to induce minimal residual disease (MRD) negativity. METHODS This academic, investigator-initiated, multicenter, phase II trial enrolled patients with high-risk NDMM (HRNDMM) defined by mandatory International Staging System stage II/III combined with del17p, t(4;14), t(14;16), or more than three 1q21 copies as high-risk cytogenetic aberrations (HRCAs). Patients received Isa-KRd induction/consolidation and Isa-KR maintenance. TE patients received high-dose melphalan. TNE patients received two additional Isa-KRd cycles postinduction. This prespecified interim analysis (IA) reports the primary end point, MRD negativity (<10-5, next-generation flow), at the end of consolidation. The secondary end point was progression-free survival (PFS). RESULTS Among 125 patients with HRNDMM (TE-intention-to-treat [ITT]-IA, 99; TNE-ITT, 26) of the IA population for the primary end point, the median age was 58 (TE-ITT-IA) and 74 (TNE-ITT) years. Del17p was the most common HRCA (TE, 44.4%; TNE, 42.3%); about one third of evaluable TE/TNE patients presented two or more HRCAs, respectively. The trial met its primary end point with MRD negativity rates after consolidation of 67.7% (TE) and 54.2% (TNE) of patients. Eighty-one of 99 TE-ITT-IA patients reached MRD negativity at any time point (81.8%). MRD negativity was sustained for ≥1 year in 62.6% of patients. With a median follow-up of 44 (TE) and 33 (TNE) months, median PFS was not reached in either arm. CONCLUSION Isa-KRd effectively induces high rates of sustainable MRD negativity in the difficult-to-treat HRNDMM population, regardless of transplant status, translating into a median PFS that was not yet reached after 44/33 months.
Collapse
Affiliation(s)
- Lisa B. Leypoldt
- Department of Hematology, Oncology and Bone Marrow Transplantation With Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Diana Tichy
- Division of Biostatistics, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Britta Besemer
- Department of Hematology, Oncology, Immunology and Rheumatology, University Hospital of Tuebingen, Tuebingen, Germany
| | - Mathias Hänel
- Department of Hematology, Oncology and Bone Marrow Transplantation, Klinikum Chemnitz, Chemnitz, Germany
| | - Marc S. Raab
- Internal Medicine V and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Mann
- Department of Hematology, Oncology and Immunology, University Hospital of Gießen and Marburg, Marburg, Germany
| | - Markus Munder
- Department of Internal Medicine III, University Medical Center Mainz, Mainz, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, German Cancer Consortium (DKTK partner site Essen), Essen, Germany
| | - Axel Nogai
- Department of Internal Medicine, Charité—University Medicine Berlin, Berlin, Germany
| | - Martin Görner
- Department of Hematology, Oncology and Palliative Care, Klinikum Bielefeld Mitte, Bielefeld, Germany
| | - Yon-Dschun Ko
- Department of Internal Medicine, Hematology and Oncology, Johanniter Krankenhaus Bonn, Bonn, Germany
| | - Maike de Wit
- Department of Internal Medicine, Hematology, Oncology and Palliative Medicine, Vivantes Klinikum Neukölln, Berlin, Germany
| | - Hans Salwender
- Asklepios Tumorzentrum Hamburg, AK Altona and AK St Georg, Hamburg, Germany
| | - Christof Scheid
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | - Ullrich Graeven
- Department of Hematology, Oncology and Gastroenterology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Rudolf Peceny
- Department of Oncology, Hematology and Stem Cell Transplantation, Klinikum Osnabrück, Osnabrück, Germany
| | - Peter Staib
- Department of Hematology and Oncology, St Antonius Hospital Eschweiler, Eschweiler, Germany
| | - Annette Dieing
- Department of Hematology and Oncology, Vivantes Klinikum am Urban, Berlin, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Michael Hundemer
- Internal Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Manola Zago
- Center for Clinical Trials, University Hospital of Tuebingen, Tuebingen, Germany
| | - Ema Požek
- Division of Biostatistics, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Axel Benner
- Division of Biostatistics, German Cancer Research Center (DKFZ) Heidelberg, Heidelberg, Germany
| | - Carsten Bokemeyer
- Department of Hematology, Oncology and Bone Marrow Transplantation With Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartmut Goldschmidt
- Internal Medicine V, GMMG-Studygroup at University Hospital Heidelberg, Heidelberg, Germany
| | - Katja C. Weisel
- Department of Hematology, Oncology and Bone Marrow Transplantation With Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
28
|
Alnasser SM, Alharbi KS, Almutairy AF, Almutairi SM, Alolayan AM. Autologous Stem Cell Transplant in Hodgkin's and Non-Hodgkin's Lymphoma, Multiple Myeloma, and AL Amyloidosis. Cells 2023; 12:2855. [PMID: 38132175 PMCID: PMC10741865 DOI: 10.3390/cells12242855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Human body cells are stem cell (SC) derivatives originating from bone marrow. Their special characteristics include their capacity to support the formation and self-repair of the cells. Cancer cells multiply uncontrollably and invade healthy tissues, making stem cell transplants a viable option for cancer patients undergoing high-dose chemotherapy (HDC). When chemotherapy is used at very high doses to eradicate all cancer cells from aggressive tumors, blood-forming cells and leukocytes are either completely or partially destroyed. Autologous stem cell transplantation (ASCT) is necessary for patients in those circumstances. The patients who undergo autologous transplants receive their own stem cells (SCs). The transplanted stem cells first come into contact with the bone marrow and then undergo engraftment, before differentiating into blood cells. ASCT is one of the most significant and innovative strategies for treating diseases. Here we focus on the treatment of Hodgkin's lymphoma, non-Hodgkin's lymphoma, multiple myeloma, and AL amyloidosis, using ASCT. This review provides a comprehensive picture of the effectiveness and the safety of ASCT as a therapeutic approach for these diseases, based on the currently available evidence.
Collapse
Affiliation(s)
- Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.F.A.)
| | - Khalid Saad Alharbi
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.F.A.)
| | - Ali F. Almutairy
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.F.A.)
| | | | | |
Collapse
|
29
|
Mo CC, Hartley-Brown MA, Midha S, Richardson PG. Upfront or Deferred Autologous Stem Cell Transplantation for Newly Diagnosed Multiple Myeloma in the Era of Triplet and Quadruplet Induction and Minimal Residual Disease/Risk-Adapted Therapy. Cancers (Basel) 2023; 15:5709. [PMID: 38136255 PMCID: PMC10741557 DOI: 10.3390/cancers15245709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
The standards of care for the initial treatment of patients with newly diagnosed multiple myeloma (NDMM) who are eligible for high-dose melphalan and autologous stem cell transplantation (HDM-ASCT) include highly active triplet and quadruplet regimens based on proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies. These regimens are resulting in improved outcomes and increasingly high rates of minimal residual disease (MRD)-negative responses without HDM-ASCT as part of the upfront therapy. Furthermore, recent randomized studies have shown that, while transplant-based approaches as a frontline therapy result in significantly longer progression-free survival compared to non-transplant approaches, this has not translated into an overall survival benefit. Given these developments, and in the context of the treatment burden of undergoing HDM-ASCT, in addition to the acute toxicities and long-term sequelae of HDM, which are associated with the genotoxicity of melphalan, there is an increasing rationale for considering deferring upfront HDM-ASCT in select transplant-eligible patients and saving it as a treatment option for later salvage therapy. Here, we review the latest clinical trial data on upfront or deferred HDM-ASCT and on the activity of quadruplet induction regimens, including rates of MRD-negative responses, and summarize emerging treatment approaches in the upfront setting such as the use of MRD-directed therapy and alternatives to HDM-ASCT.
Collapse
Affiliation(s)
| | | | | | - Paul G. Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, 450 Brookline Avenue, Dana 1B02, Boston, MA 02115, USA; (C.C.M.); (M.A.H.-B.); (S.M.)
| |
Collapse
|
30
|
Ziccheddu B, Giannotta C, D'Agostino M, Bertuglia G, Saraci E, Oliva S, Genuardi E, Papadimitriou M, Diamond B, Corradini P, Coffey D, Landgren O, Bolli N, Bruno B, Boccadoro M, Massaia M, Maura F, Larocca A. Genomic and immune determinants of resistance to anti-CD38 monoclonal antibody-based therapy in relapsed refractory multiple myeloma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23299287. [PMID: 38106151 PMCID: PMC10723485 DOI: 10.1101/2023.12.04.23299287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Anti-CD38 antibody therapies have transformed multiple myeloma (MM) treatment. However, a large fraction of patients inevitably relapses. To understand this, we investigated 32 relapsed MM patients treated with daratumumab, lenalidomide, and dexamethasone (Dara-Rd; NCT03848676 ). Whole genome sequencing (WGS) before and after treatment pinpointed genomic drivers associated with early progression, including RPL5 loss and APOBEC mutagenesis. Flow cytometry on 202 blood samples, collected every three months until progression for 31 patients, revealed distinct immune changes significantly impacting clinical outcomes. Progressing patients exhibited significant depletion of CD38+ NK cells, persistence of T cell exhaustion, and reduced depletion of T-reg cells over time. These findings underscore the influence of immune composition and daratumumab-induced immune changes in promoting MM resistance. Integrating genomics and flow cytometry unveiled associations between adverse genomic features and immune patterns. Overall, this study sheds light on the intricate interplay between genomic complexity and the immune microenvironment driving resistance to Dara-Rd.
Collapse
|
31
|
Maura F, Boyle EM, Coffey D, Maclachlan K, Gagler D, Diamond B, Ghamlouch H, Blaney P, Ziccheddu B, Cirrincione A, Chojnacka M, Wang Y, Siegel A, Hoffman JE, Kazandjian D, Hassoun H, Guzman E, Mailankody S, Shah UA, Tan C, Hultcrantz M, Scordo M, Shah GL, Landau H, Chung DJ, Giralt S, Zhang Y, Arbini A, Gao Q, Roshal M, Dogan A, Lesokhin AM, Davies FE, Usmani SZ, Korde N, Morgan GJ, Landgren O. Genomic and immune signatures predict clinical outcome in newly diagnosed multiple myeloma treated with immunotherapy regimens. NATURE CANCER 2023; 4:1660-1674. [PMID: 37945755 PMCID: PMC12065606 DOI: 10.1038/s43018-023-00657-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 09/20/2023] [Indexed: 11/12/2023]
Abstract
Despite improving outcomes, 40% of patients with newly diagnosed multiple myeloma treated with regimens containing daratumumab, a CD38-targeted monoclonal antibody, progress prematurely. By integrating tumor whole-genome and microenvironment single-cell RNA sequencing from upfront phase 2 trials using carfilzomib, lenalidomide and dexamethasone with daratumumab ( NCT03290950 ), we show how distinct genomic drivers including high APOBEC mutational activity, IKZF3 and RPL5 deletions and 8q gain affect clinical outcomes. Furthermore, evaluation of paired bone marrow profiles, taken before and after eight cycles of carfilzomib, lenalidomide and dexamethasone with daratumumab, shows that numbers of natural killer cells before treatment, high T cell receptor diversity before treatment, the disappearance of sustained immune activation (that is, B cells and T cells) and monocyte expansion over time are all predictive of sustained minimal residual disease negativity. Overall, this study provides strong evidence of a complex interplay between tumor cells and the immune microenvironment that is predictive of clinical outcome and depth of treatment response in patients with newly diagnosed multiple myeloma treated with highly effective combinations containing anti-CD38 antibodies.
Collapse
Affiliation(s)
- Francesco Maura
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
| | - Eileen M Boyle
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - David Coffey
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Dylan Gagler
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Benjamin Diamond
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Hussein Ghamlouch
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Patrick Blaney
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Bachisio Ziccheddu
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Anthony Cirrincione
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Monika Chojnacka
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Yubao Wang
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Ariel Siegel
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - James E Hoffman
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Dickran Kazandjian
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emily Guzman
- Genome Technology Center, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Urvi A Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Carlyn Tan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Michael Scordo
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunjan L Shah
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heather Landau
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David J Chung
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio Giralt
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yanming Zhang
- Cytogenetics Laboratory, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnaldo Arbini
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Qi Gao
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmet Dogan
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander M Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Faith E Davies
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA
| | - Saad Z Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Gareth J Morgan
- Myeloma Research Program, NYU Langone, Perlmutter Cancer Center, New York, NY, USA.
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
32
|
Bar N, Firestone RS, Usmani SZ. Aiming for the cure in myeloma: Putting our best foot forward. Blood Rev 2023; 62:101116. [PMID: 37596172 DOI: 10.1016/j.blre.2023.101116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/20/2023]
Abstract
Frontline therapy for multiple myeloma (MM) is evolving to include novel combinations that can achieve unprecedented deep response rates. Several treatment strategies exist, varying in induction regimen composition, use of transplant and or consolidation and maintenance. In this sea of different treatment permutations, the overarching theme is the powerful prognostic factors of disease risk and achievement of minimal residual disease (MRD) negativity. MM has significant inter-patient variability that requires treatment to be individualized. Risk-adapted and response-adapted strategies which are increasingly being explored to define the extent and duration of therapy, and eventually aim for functional curability. In addition, with T-cell redirection therapies rapidly revolutionizing myeloma treatments, the current standard of care for myeloma will change. This review analyzes the current relevant literature in upfront therapy for fit myeloma patients and provides suggestions for treatment approach while novel clinical trials are maturing.
Collapse
Affiliation(s)
- Noffar Bar
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine University, New Haven, CT, USA.
| | - Ross S Firestone
- Multiple Myeloma Service, Department of medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Saad Z Usmani
- Multiple Myeloma Service, Department of medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
33
|
Costa LJ, Chhabra S, Medvedova E, Dholaria BR, Schmidt TM, Godby KN, Silbermann R, Dhakal B, Bal S, Giri S, D'Souza A, Hall AC, Hardwick P, Omel J, Cornell RF, Hari P, Callander NS. Minimal residual disease response-adapted therapy in newly diagnosed multiple myeloma (MASTER): final report of the multicentre, single-arm, phase 2 trial. Lancet Haematol 2023; 10:e890-e901. [PMID: 37776872 PMCID: PMC10836587 DOI: 10.1016/s2352-3026(23)00236-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/22/2023] [Accepted: 07/27/2023] [Indexed: 10/02/2023]
Abstract
BACKGROUND For patients with newly diagnosed multiple myeloma, reaching minimal residual disease (MRD) negativity after treatment is associated with improved outcomes; however, the use of MRD to modulate therapy remains elusive. We present the final analysis of the MASTER trial of daratumumab, carfilzomib, lenalidomide, and dexamethasone (Dara-KRd) therapy in patients with newly diagnosed multiple myeloma, in which MRD status is used to modulate treatment duration and cessation. METHODS MASTER was a multicentre, single-arm, phase 2 trial conducted in five academic medical centres in the USA. Eligible participants were 18 years or older with newly diagnosed multiple myeloma (measurable by serum or urine protein electrophoresis or serum free light chains), a life expectancy of at least 12 months, and an Eastern Cooperative Oncology Group performance status of 0-2, and had received no previous treatment for multiple myeloma except up to one cycle of therapy containing bortezomib, cyclophosphamide, and dexamethasone. The study was enriched for participants with high-risk chromosome abnormalities (HRCAs). During the induction phase, participants received four 28-day cycles of Dara-KRd, each comprising daratumumab (16 mg/kg intravenously on days 1, 8, 15, and 22), carfilzomib (56 mg/m2 intravenously on days 1, 8, and 15), lenalidomide (25 mg orally on days 1-21), and dexamethasone (40 mg orally or intravenously on days 1, 8, 15, and 22); induction was followed by autologous haematopoietic stem-cell transplantation and up to two phases of consolidation with Dara-KRd. We assessed MRD by next-generation sequencing after or during each phase. The primary endpoint was reaching MRD negativity (<10-5). Participants who reached MRD negativity after or during two consecutive phases stopped treatment and began observation with MRD surveillance (MRD-SURE); participants who did not reach two consecutive MRD-negative results received maintenance lenalidomide. Secondary endpoints included progression-free survival and cumulative incidence of progression. All analyses were conducted in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, NCT03224507, and is complete. FINDINGS Between Mar 21, 2018, and Oct 23, 2020, 123 participants were recruited to the study, of whom 70 (57%) were men, 53 (43%) were women, 94 (76%) were non-Hispanic White, 25 (20%) were non-Hispanic Black, and four (3%) were of another race or ethnicity. The median age of participants was 61 years (IQR 55-68), and 24 (20%) were aged 70 years or older. The median duration of follow up was 42·2 months (IQR 34·5-46·0). Of the 123 participants, 53 (43%) had no HRCAs, 46 (37%) had one HRCA, and 24 (20%) had two or more HRCAs. For 118 (96%) of 123 participants, MRD was evaluable by next-generation sequencing; the remaining five had an absence of sufficiently unique clonogenic sequences to enable tracking by the assay. Of these 118 participants, 96 (81%, 95% CI 73-88) reached MRD of less than 10-5 (comprising 39 [78%, 64-88] of 50 participants with no HRCAs, 38 [86%, 73-95] of 44 participants with one HRCA, and 19 [79%, 58-93] of 24 participants with two or more HRCAs) and 84 (71%, 62-79) reached MRD-SURE and treatment cessation. 36-month progression-free survival among all 123 participants was 88% (95% CI 78-95) for participants with no HRCAs, 79% (67-88) for those with one HRCA, and 50% (30-70) for those with two or more HRCAs. For the 84 participants reaching MRD-SURE, the 24-month cumulative incidence of progression from cessation of therapy was 9% (95% CI 1-19) for participants with no HRCAs, 9% (1-18) for those with one HRCA, and 47% (23-72) for those with two or more HRCAs. 61 participants (comprising 52% of 118 MRD-evaluable participants and 73% of 84 participants who reached MRD-SURE) remain free of therapy and MRD-negative as of Feb 7, 2023. The most common grade 3-4 adverse events were neutropenia (43 patients, 35%), lymphopenia (28 patients, 23%), and hypertension (13 patients, 11%). Three treatment-emergent deaths were recorded: two sudden deaths and one due to viral infection, none of which were judged to be treatment-related. INTERPRETATION This approach provided positive outcomes and a pathway for treatment cessation in most patients with newly diagnosed multiple myeloma. Outcomes for patients with ultra-high-risk multiple myeloma, defined as those with two or more HRCAs, remain unsatisfactory, and these patients should be prioritised for trials with early introduction of therapies with novel mechanisms of action. FUNDING Amgen and Janssen Pharmaceuticals.
Collapse
Affiliation(s)
- Luciano J Costa
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Saurabh Chhabra
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva Medvedova
- Division of Hematology and Medical Oncology, Department of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Bhagirathbhai R Dholaria
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy M Schmidt
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of Wisc onsin, Madison, WI, USA
| | - Kelly N Godby
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rebecca Silbermann
- Division of Hematology and Medical Oncology, Department of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Binod Dhakal
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Susan Bal
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Smith Giri
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anita D'Souza
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aric C Hall
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of Wisc onsin, Madison, WI, USA
| | - Pamela Hardwick
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - James Omel
- Academic Consortium to Overcome Multiple Myeloma through Innovative Trials (COMMIT), Omaha, NE, USA
| | - Robert F Cornell
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Parameswaran Hari
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Natalie S Callander
- Division of Hematology, Oncology and Palliative Care, Department of Medicine, University of Wisc onsin, Madison, WI, USA
| |
Collapse
|
34
|
Derman BA, Zonder J, Reece D, Cole C, Berdeja J, Stefka AT, Major A, Kin A, Griffith K, Jasielec J, Jakubowiak AJ. Phase 1/2 study of carfilzomib, pomalidomide, and dexamethasone with and without daratumumab in relapsed multiple myeloma. Blood Adv 2023; 7:5703-5712. [PMID: 36763537 PMCID: PMC10539870 DOI: 10.1182/bloodadvances.2022008866] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 02/11/2023] Open
Abstract
We conducted a phase 1/2 study of carfilzomib, pomalidomide, and dexamethasone (KPd) and KPd with daratumumab (Dara-KPd) in relapsed/refractory multiple myeloma. The primary end points were identification of a maximum tolerated dose (MTD) of KPd for phase 1, and rates of overall response (ORR) and near complete response (nCR) after 4 cycles of KPd and Dara-KPd, respectively, for phase 2. The MTD for KPd was carfilzomib 20/27 mg/m2 on days 1, 2, 8, 9, 15, and 16 (cycles 1-8) and days 1, 2, 15, and 16 for cycles 9 and beyond; oral pomalidomide 4 mg on days 1 to 21; and oral dexamethasone 40 mg weekly in 28-day cycles. Sixty-six patients received KPd, including 34 at the MTD. The ORR after 4 cycles of KPd at the MTD was 27/34 (79%; 95% confidence interval [CI], 62%-91%), meeting the statistical threshold for efficacy. At a median follow-up of 44 months, the median progression-free survival (PFS) was 13 months and overall survival (OS) 44 months. Twenty-eight patients received Dara-KPd. The rate of nCR or better after 4 cycles was 11/28 (39%; 95% CI, 22%-59%), meeting the statistical threshold for efficacy. As the best response to Dara-KPd, the ORR was 25/28 (89%) and the rate of measurable residual disease negativity by flow cytometry (10-5) was 17/26 (65%). At a median follow-up of 26 months, the median PFS and OS for Dara-KPd were not reached. Dara-KPd induced deeper and more durable responses than KPd without compromising safety in a predominantly high-risk, lenalidomide-refractory population, warranting further evaluation of this quadruplet. This trial is registered at www.clinicaltrials.gov as #NCT01665794.
Collapse
Affiliation(s)
| | - Jeffrey Zonder
- Karmanos Cancer Center, Wayne State University, Detroit, MI
| | - Donna Reece
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Craig Cole
- Michigan State University, East Lansing, MI
- University of Michigan, Ann Arbor, MI
| | | | - Andrew T. Stefka
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Ajay Major
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | - Andrew Kin
- Karmanos Cancer Center, Wayne State University, Detroit, MI
| | | | - Jagoda Jasielec
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
- Janssen Pharmaceuticals, Titusville, NJ
| | | |
Collapse
|
35
|
Abstract
SUMMARY In this issue, Paiva and colleagues characterize the dynamics of minimal residual disease (MRD) and clinical responses during chimeric antigen receptor (CAR) T-cell therapy of relapsed/refractory multiple myeloma. Although both correlate with prolonged progression-free survival, MRD is reached faster in the bone marrow than complete response in peripheral blood; consequently, the study addresses the need for future guidelines to explore new MRD-negative definitions that are independent of the monoclonal (M) protein to overcome this limitation, particularly in clinical trials using early depth of response as an endpoint. See related article by Paiva et al., p. 365 (1).
Collapse
Affiliation(s)
- Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Dickran Kazandjian
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
36
|
Kegyes D, Gulei D, Drula R, Cenariu D, Tigu B, Dima D, Tanase A, Badelita S, Buzoianu AD, Ciurea S, Ghiaur G, Terpos E, Ciechanover A, Einsele H, Tomuleasa C. Proteasome inhibition in combination with immunotherapies: State-of-the-Art in multiple myeloma. Blood Rev 2023; 61:101100. [PMID: 37291017 DOI: 10.1016/j.blre.2023.101100] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder accounting for around 1.8% of all neoplastic diseases. Nowadays, clinicians have a broad arsenal of drugs at their disposal for the treatment of MM, such as proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, bispecific antibodies, CAR T-cell therapies and antibody-drug conjugates. In this paper we briefly highlight essential clinical elements relating to proteasome inhibitors, such as bortezomib, carfilzomib and ixazomib. Studies suggest that the early use of immunotherapy may improve outcomes significantly. Therefore, in our review we specifically focus on the combination therapy of proteasome inhibitors with novel immunotherapies and/or transplant. A high number of patients develop PI resistance. Thus, we also review new generation PIs, such as marizomib, oprozomib (ONX0912) and delanzomib (CEP-18770) and their combinations with immunotherapies.
Collapse
Affiliation(s)
- David Kegyes
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Rares Drula
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Diana Cenariu
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bogdan Tigu
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Alina Tanase
- Department of Hematology and Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Sorina Badelita
- Department of Hematology and Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Anca-Dana Buzoianu
- Department of Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Stefan Ciurea
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California Irvine, CA, United States
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Aaron Ciechanover
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hermann Einsele
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
| |
Collapse
|
37
|
Chakraborty R, Al Hadidi S, Scheffer Cliff ER, Mohyuddin GR. Is aggressive treatment of smoldering myeloma the path to curing myeloma? Blood Adv 2023; 7:3932-3935. [PMID: 37196639 PMCID: PMC10405196 DOI: 10.1182/bloodadvances.2023009658] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Affiliation(s)
- Rajshekhar Chakraborty
- Multiple Myeloma and Amyloidosis Program, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Samer Al Hadidi
- Myeloma Section, Winthrop P. Rockefeller Cancer Institute at the University of Arkansas for Medical Sciences, Little Rock, AR
| | - Edward R. Scheffer Cliff
- Program on Regulation, Therapeutics and Law, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
38
|
Oriol A, Abril L, Ibarra G. First-line treatment of multiple myeloma in both transplant and non-transplant candidates. Expert Rev Anticancer Ther 2023; 23:685-698. [PMID: 37194283 DOI: 10.1080/14737140.2023.2213891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023]
Abstract
INTRODUCTION The last decade's progress in the treatment of newly diagnosed multiple myeloma has relied on the synergistic combination of agents with different mechanisms of action, basically proteasome inhibitors, immunomodulatory agents, and monoclonal antibodies, in order to achieve the deepest possible response early in the course of treatment. Following induction, several therapeutic strategies aim to improve and maintain response. AREAS COVERED The manuscript reviews available data for the treatment of newly diagnosed multiple myeloma patients with a focus on most recent induction and maintenance combinations and the still important role of autologous stem transplantation. Future perspectives in the light of initial results from ongoing clinical trials are also addressed. EXPERT OPINION Remarkable progress has been made in myeloma treatment due to the integration of immunomodulators, proteasome inhibitors, monoclonal antibodies, and high dose therapy in the frontline setting. Upfront therapy may be further improved intensifying induction combinations, adapting high dose therapy and consolidation strategies to the patient's profile, improving maintenance in high-risk individuals, or limiting maintenance duration in those with a better prognosis. Evidence needs to be reviewed, taking into account the therapeutic objectives at each treatment stage and patient specific risk factors.
Collapse
Affiliation(s)
- Albert Oriol
- Institut Català d'Oncologia and Institut Josep Carreras. Hospital Germans Trias i Pujol, Carretera del Canyet, Barcelona, Spain
| | - Laura Abril
- Institut Català d'Oncologia and Institut Josep Carreras. Hospital Germans Trias i Pujol, Carretera del Canyet, Barcelona, Spain
| | - Gladys Ibarra
- Institut Català d'Oncologia and Institut Josep Carreras. Hospital Germans Trias i Pujol, Carretera del Canyet, Barcelona, Spain
| |
Collapse
|
39
|
Tanguay M, Dagenais C, LeBlanc R, Ahmad I, Claveau JS, Roy J. Young Myeloma Patients: A Systematic Review of Manifestations and Outcomes. Curr Oncol 2023; 30:5214-5226. [PMID: 37366879 DOI: 10.3390/curroncol30060396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Multiple myeloma usually affects older adults. However, younger patients constitute a significant subset as approximately 10% of cases occur in subjects younger than 50 years old. Young patients, who are underrepresented in the literature, are diagnosed during their most productive years of life, urging the need for tailored treatment approaches. This literature review aims to report recent studies specifically addressing young patients with a focus on characteristics at diagnosis, cytogenetics, treatments, and outcomes. We searched PubMed for studies involving young patients with multiple myeloma ≤50 years old. The time span of our literature review search was from 1 January 2010 to 31 December 2022. Overall, 16 retrospective studies were analyzed for this review. Young patients with multiple myeloma tend to have less advanced disease, more frequent light chain subtypes, and survive longer compared to their older counterparts. However, available studies included a limited number of patients; the newest revised international staging system was not used to stratify patients, cytogenetics varied from one cohort to another, and most patients did not receive contemporary triplet/quadruplet treatments. This review emphasizes the need to perform contemporary, large-scale retrospective studies to improve knowledge regarding the presentation and outcomes of young myeloma patients in the era of modern treatments.
Collapse
Affiliation(s)
- Mégane Tanguay
- Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, 5415 de l'Assomption, Montreal, QC H1T 2M4, Canada
- Departement of Medicine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Christophe Dagenais
- Departement of Medicine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Service de Médecine Interne, Hôpital Notre-Dame, 1560 Sherbrooke Est, Montreal, QC H2L 4M1, Canada
| | - Richard LeBlanc
- Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, 5415 de l'Assomption, Montreal, QC H1T 2M4, Canada
- Departement of Medicine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Imran Ahmad
- Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, 5415 de l'Assomption, Montreal, QC H1T 2M4, Canada
- Departement of Medicine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Jean-Sébastien Claveau
- Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, 5415 de l'Assomption, Montreal, QC H1T 2M4, Canada
- Departement of Medicine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Jean Roy
- Institut Universitaire d'Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, 5415 de l'Assomption, Montreal, QC H1T 2M4, Canada
- Departement of Medicine, Université de Montréal, 2900 Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
40
|
何 旎, 周 文. [Latest Findings on the Mechanism of the Interaction Between Multiple Myeloma Cells and Bone Marrow Microenvironment]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:475-481. [PMID: 37248571 PMCID: PMC10475439 DOI: 10.12182/20230560207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Indexed: 05/31/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy of terminally differentiated plasma cells. The mechanisms of the pathogenesis and progression of MM include genetic abnormalities of the MM cells and the interaction between MM cells and bone marrow microenvironment (BMME). MM cells start malignant proliferation in BMME and contribute to the pathogenesis and progression of MM through direct or indirect interactions between cells and the extracellular matrix. Exploring the mechanism of interaction between MM cells and the microenvironment is crucial to improving our understanding of the pathogenesis and progression of MM and early diagnosis and treatment. In addition, the metabolic reprogramming of tumors is one of the key issues of oncology research. Herein, we summarized published findings on the the altered metabolic reprogramming of MM and the characteristics of MM metabolic-microbial interactions in order to gain an in-depth understanding of MM pathogenesis and progression and drug resistance mechanisms, and ultimately to explore for new strategies for MM treatment.
Collapse
Affiliation(s)
- 旎涵 何
- 中南大学基础医学院 肿瘤研究所 (长沙 412000)Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 412000, China
| | - 文 周
- 中南大学基础医学院 肿瘤研究所 (长沙 412000)Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 412000, China
| |
Collapse
|
41
|
Morè S, Corvatta L, Manieri VM, Olivieri A, Offidani M. Current Main Topics in Multiple Myeloma. Cancers (Basel) 2023; 15:2203. [PMID: 37190132 PMCID: PMC10136770 DOI: 10.3390/cancers15082203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Multiple Myeloma (MM) remains a difficult to treat disease mainly due to its biological heterogeneity, of which we are more and more knowledgeable thanks to the development of increasingly sensitive molecular methods that allow us to build better prognostication models. The biological diversity translates into a wide range of clinical outcomes from long-lasting remission in some patients to very early relapse in others. In NDMM transplant eligible (TE) patients, the incorporation of mAb as daratumumab in the induction regimens, followed by autologous stem cell transplantation (ASCT) and consolidation/maintenance therapy, has led to a significant improvement of PFS and OS.; however, this outcome remains poor in ultra-high risk MM or in those who did not achieve a minimal residual disease (MRD) negativity. Several trials are exploring cytogenetic risk-adapted and MRD-driven therapies in these patients. Similarly, quadruplets-containing daratumumab, particularly when administered as continuous therapies, have improved outcome of patients not eligible for autologous transplant (NTE). Patients who become refractory to conventional therapies have noticeably poor outcomes, making their treatment a difficult challenge in need of novel strategies. In this review, we will focus on the main points regarding risk stratification, treatment and monitoring of MM, highlighting the most recent evidence that could modify the management of this still incurable disease.
Collapse
Affiliation(s)
- Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| | - Laura Corvatta
- Unità Operativa Complessa di Medicina, Ospedale Profili, 60044 Fabriano, Italy
| | | | - Attilio Olivieri
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| | - Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| |
Collapse
|
42
|
Charalampous C, Goel U, Gertz M, Lacy M, Dispenzieri A, Hayman S, Dingli D, Buadi F, Kapoor P, Kourelis T, Warsame R, Hogan WJ, Kumar S. Impact of the time interval between end of induction and autologous hematopoietic transplantation in newly diagnosed patients with multiple myeloma. Bone Marrow Transplant 2023; 58:46-53. [PMID: 36203088 PMCID: PMC9812760 DOI: 10.1038/s41409-022-01835-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 01/10/2023]
Abstract
Multiple Myeloma patients eligible for autologous hematopoietic transplantation (AHT) typically receive 3-6 cycles of induction therapy before transplant. The last induction cycle is completed 2-4 weeks prior to mobilization. We evaluated the impact of the time interval between end of induction and AHT on progression-free survival (PFS) and overall survival (OS). A total of 1055 patients who underwent AHT were identified. The median time to transplant (TTT) was 33 days (27-42 quartile range). Patients with less than 33 days of TTT had significantly prolonged PFS (35.6 vs. 32.1 months, p < 0.03) but non-significant OS differences compared to those with more than 33 days. Quartile comparisons showed that patients in the 1st quartile (less than 27 days) had significantly prolonged PFS (36.7 vs. 30.9 months, p < 0.01) compared to the 4th quartile group (more than 42 days). In a subgroup analysis of patients with partial or worse biochemical response prior to transplant, patients in the 1st quartile had significantly prolonged PFS (37.7 vs. 28.7 months, p < 0.04) compared to the 4th quartile group. In conclusion, we showed that a prolonged TTT is associated with inferior outcomes compared to tighter chemotherapy schedules. This finding was especially prevalent in patients with partial response at induction.
Collapse
Affiliation(s)
- Charalampos Charalampous
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Utkarsh Goel
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Morie Gertz
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Martha Lacy
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Angela Dispenzieri
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Suzanne Hayman
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - David Dingli
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Francis Buadi
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Prashant Kapoor
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Taxiarchis Kourelis
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Rahma Warsame
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - William J. Hogan
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN USA
| | - Shaji Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
43
|
Costello CL. Newly diagnosed multiple myeloma: making sense of the menu. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:539-550. [PMID: 36485145 PMCID: PMC9820388 DOI: 10.1182/hematology.2022000404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The development of new drugs and subsequent novel combinations for the treatment of newly diagnosed multiple myeloma (NDMM) has resulted in a plethora of treatment options that can make the choice of initial induction therapy a challenge. A greater understanding of both patient- and disease-specific factors can provide a personalized approach to help design a treatment course. Historically, the choice of an induction regimen has been tethered to an initial impression of transplant eligibility at the time of diagnosis. As more effective and better-tolerated induction regimens have emerged, there has been increasing overlap in the induction strategies used for all patients with NDMM, which increasingly provide the ultimate goal of deep and durable remissions. The current treatment options and strategies for the management of NDMM are evaluated using the best available data to provide a rationale for these decisions.
Collapse
Affiliation(s)
- Caitlin L. Costello
- Correspondence Caitlin L. Costello, UCSD, 3855 Health Sciences Drive, MC 0960, La Jolla, CA 92093, USA; e-mail:
| |
Collapse
|
44
|
Cipkar C, Chen C, Trudel S. Antibodies and bispecifics for multiple myeloma: effective effector therapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:163-172. [PMID: 36485135 PMCID: PMC9820318 DOI: 10.1182/hematology.2022000334] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The therapeutic landscape in multiple myeloma (MM) has changed dramatically over the last 2 decades. With the introduction of novel immunotherapies, patients with MM can expect deeper responses, longer remissions, and improved overall survival. Since its approval by the US Food and Drug Administration in 2015, the monoclonal antibody specific for CD38, daratumumab, has been incorporated into both frontline and relapsed treatment regimens. Its role as a maintenance therapy is currently being explored. Subsequently, a variety of novel antibody therapeutics have evolved from the success of daratumumab, using similar concepts to target the malignant plasma cell clone. Noteworthy naked monoclonal antibodies include isatuximab, another agent directed against CD38, and elotuzumab, an agent directed against SLAM family member 7. Antibody-drug conjugates, complex molecules composed of an antibody tethered to a cytotoxic drug, target malignant cells and deliver a lethal payload. The first to market is belantamab mafodotin, which targets B-cell maturation antigen (BCMA) on malignant plasma cells and delivers a potent microtubule inhibitor, monomethyl auristatin F. Additionally, bispecific T-cell antibodies are in development that engage the immune system directly by simultaneously binding CD3 on T cells and a target epitope-such as BCMA, G-protein coupled receptor family C group 5 member D (GPRC5d), and Fc receptor homologue 5 (FcRH5)-on malignant cells. Currently, teclistamab, an anti-BCMA bispecific, is closest to approval for commercial use. In this review, we explore the evolving landscape of antibodies in the treatment of MM, including their role in frontline and relapse settings.
Collapse
Affiliation(s)
- Christopher Cipkar
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Christine Chen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Suzanne Trudel
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
45
|
Nath K, Shekarkhand T, Salcedo M, Derkach A, Rueda S, Chansakul A, Hulcrantz M, Korde N, Shah UA, Tan C, Chung DJ, Lahoud OB, Hassoun H, Lesokhin AM, Landau HJ, Shah G, Scordo M, Giralt SA, Usmani SZ, Roshal M, Landgren O, Mailankody S. A short course of daratumumab in patients with multiple myeloma and minimal residual disease after induction therapy. Leuk Lymphoma 2022; 63:3488-3492. [PMID: 36282633 DOI: 10.1080/10428194.2022.2131417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 01/25/2023]
Affiliation(s)
- Karthik Nath
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tala Shekarkhand
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meghan Salcedo
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andriy Derkach
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siobhan Rueda
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Aisara Chansakul
- Emergency Department, Division of Emergency Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Malin Hulcrantz
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neha Korde
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Urvi A Shah
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carlyn Tan
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David J Chung
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oscar B Lahoud
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hani Hassoun
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander M Lesokhin
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heather J Landau
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunjan Shah
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Scordo
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio A Giralt
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Saad Z Usmani
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Sham Mailankody
- Department of Medicine, Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
46
|
Kazandjian D, Kowalski A, Landgren O. T cell redirecting bispecific antibodies for multiple myeloma: emerging therapeutic strategies in a changing treatment landscape. Leuk Lymphoma 2022; 63:3032-3043. [PMID: 36059239 PMCID: PMC10113039 DOI: 10.1080/10428194.2022.2113532] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/20/2022] [Accepted: 08/02/2022] [Indexed: 01/11/2023]
Abstract
In recent years, the treatment landscape of multiple myeloma has continued to evolve with the introduction of novel immunotherapies. This progress has translated to improved overall survival for patients, but an unmet need remains in the heavily pretreated and high-risk subsets of patients. Emerging immunotherapies in the form of CAR-T cell therapies have been approved for multiple myeloma. However, CAR-T cell therapy has logistical limitations and there is a need for immunotherapies that are readily available, safe, and effective in RRMM. Currently, pending approval, there are many "off the shelf" bispecific antibodies being developed that target BCMA, GPRC5D, FcRH5 and other cell surface proteins. Preliminary efficacy data has suggested that these bispecific antibody therapies have similar response rates (∼50-80%) in heavily pretreated patients. Similarly, to CAR-T cell therapy, cytokine release syndrome and immune effector cell associated neurotoxicity syndrome are adverse events of key interest and incidence range from ∼40 to 90% and 3 to 20%, respectively. In this review, we highlight the various bispecific immunotherapies under development in the treatment of multiple myeloma with a focus on the data from clinical phase I and II studies.
Collapse
Affiliation(s)
- Dickran Kazandjian
- Myeloma Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami
| | - Andrew Kowalski
- Myeloma Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami
| | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami
| |
Collapse
|
47
|
Goldsmith SR, Ghobadi A, Dipersio JF, Hill B, Shadman M, Jain T. Chimeric Antigen Receptor T Cell Therapy versus Hematopoietic Stem Cell Transplantation: An Evolving Perspective. Transplant Cell Ther 2022; 28:727-736. [PMID: 35878743 PMCID: PMC10487280 DOI: 10.1016/j.jtct.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/30/2022] [Accepted: 07/16/2022] [Indexed: 11/27/2022]
Abstract
Cellular therapy modalities, including autologous (auto-) hematopoietic cell transplantation (HCT), allogeneic (allo-) HCT, and now chimeric antigen receptor (CAR) T cell therapy, have demonstrated long-term remission in advanced hematologic malignancies. Auto-HCT and allo-HCT, through hematopoietic rescue, have permitted the use of higher doses of chemotherapy. Allo-HCT also introduced a nonspecific immune-mediated targeting of malignancy resulting in protection from relapse, although at the expense of similar targeting of normal host cells. In contrast, CAR T therapy, through genetically engineered immunotherapeutic precision, allows for redirection of autologous immune effector cells against malignancy in an antigen-specific and MHC-independent fashion, with demonstrated efficacy in patients who are refractory to cytotoxic chemotherapy. It too has unique toxicities and challenges, however. Non-Hodgkin lymphoma (including large B cell lymphoma, mantle cell lymphoma, and follicular lymphoma), B cell acute lymphoblastic leukemia, and multiple myeloma are the 3 main diseases associated with the use of fully developed CAR T products with widespread deployment. Recent and ongoing clinical trials have been examining the interface among the 3 cellular therapy modalities (auto-HCT, allo-HCT, and CAR T) to determine whether they should be "complementary" or "competitive" therapies. In this review, we examine the current state of this interface with respect to the most recent data and delve into the controversies and conclusions that may inform clinical decision making.
Collapse
Affiliation(s)
- Scott R Goldsmith
- Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope Comprehensive Cancer Center, Duarte, California; Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri.
| | - Armin Ghobadi
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - John F Dipersio
- Division of Oncology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Brian Hill
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Mayzar Shadman
- Clinical Research Division, Fred Hutch Cancer Center and Medical Oncology division, University of Washington, Seattle, Washington
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
48
|
Goel U, Kumar S. An update on the safety of ixazomib for the treatment of multiple myeloma. Expert Opin Drug Saf 2022; 21:1143-1160. [PMID: 36178708 DOI: 10.1080/14740338.2022.2130892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Treatment options for multiple myeloma (MM) have rapidly expanded over the past few years with several newly approved drugs. While there is need to explore treatments that lead to longer responses and survival, special consideration should be given on reducing treatment burden, reducing toxicities, and improving quality of life. Ixazomib is the first oral proteasome inhibitor for the treatment of MM, combining clinical efficacy with a favorable safety profile. AREAS COVERED Here, we discuss the clinical efficacy and safety of ixazomib. Pharmacokinetic considerations, management of common toxicities, and the impact of the drug on the current and future treatment strategies are also discussed. EXPERT OPINION Ixazomib is an effective and welltolerated MM drug. It is also being studied in combination with other newer agents. It does not have long-term cumulative toxicities, and the most adverse events are mild and manageable. These findings, along with the ease of oral administration, make it a possible option for long-term treatment approaches for MM patients, as well as in the frail/elderly patient population.
Collapse
Affiliation(s)
- Utkarsh Goel
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Shaji Kumar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
49
|
McCaughan GJ, Gandolfi S, Moore JJ, Richardson PG. Lenalidomide, bortezomib and dexamethasone induction therapy for the treatment of newly diagnosed multiple myeloma: a practical review. Br J Haematol 2022; 199:190-204. [PMID: 35796524 PMCID: PMC9796722 DOI: 10.1111/bjh.18295] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 01/07/2023]
Abstract
For patients with newly diagnosed multiple myeloma, survival outcomes continue to improve significantly: however, nearly all patients will relapse following induction treatment. Optimisation of induction therapy is essential to provide longer term disease control and the current standard of care for most patients incorporates an immunomodulatory agent and proteasome inhibitor, most commonly lenalidomide and bortezomib in combination with dexamethasone (RVD), with maintenance until progression. Historically there has been limited access to RVD as an induction strategy outside of the United States; fortunately, there is now increasing access worldwide. This review discusses the rationale for use of RVD as induction therapy and aims to provide guidance in prescribing this regimen in order to optimise efficacy while minimising the toxicities of treatment. We also highlight the increasing evidence for the utility of addition of a monoclonal antibody to the RVD backbone to deepen responses and potentially provide longer disease control.
Collapse
Affiliation(s)
- Georgia J. McCaughan
- Department of HaematologySt Vincent's HospitalSydneyAustralia
- University of New South Wales, Medicine and HealthSydneyAustralia
| | - Sara Gandolfi
- Translational Research ProgramUniversity of HelsinkiHelsinkiFinland
- Haematology Research UnitUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - John J. Moore
- Department of HaematologySt Vincent's HospitalSydneyAustralia
- University of New South Wales, Medicine and HealthSydneyAustralia
| | - Paul G. Richardson
- Dana‐Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Department of Medical OncologyBostonMassachusettsUSA
| |
Collapse
|
50
|
Landgren O, Kazandjian D, Roussel M, Jasielec J, Dytfeld D, Anderson A, Kervin TA, Iskander K, McFadden I, Jakubowiak AJ. Efficacy and safety of carfilzomib-lenalidomide-dexamethasone in newly diagnosed multiple myeloma: pooled analysis of four single-arm studies. Leuk Lymphoma 2022; 63:2413-2421. [PMID: 35549810 DOI: 10.1080/10428194.2022.2068001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 10/18/2022]
Abstract
Pooled analyses of four single-arm phase 1 and 2 studies (NCT01816971, NCT02405364, NCT01029054, NCT01402284) investigated the clinical effectiveness of carfilzomib-lenalidomide-dexamethasone (KRd) in newly diagnosed multiple myeloma (NDMM). Patients who did (Cohort 1; n = 122) and did not (Cohort 2; n = 99) undergo autologous stem cell transplant (high-dose melphalan [HDM]-ASCT) were included. Patients received a 28-day cycle of induction KRd. The rate of very good partial response or better, the primary endpoint, was 93% in Cohort 1 and 90% in Cohort 2. Two-year progression-free survival and overall survival rates were 88% and 96% for Cohort 1, and 85% and 97% for Cohort 2. At least 90% of patients in each cohort reported ≥1 grade 3 or 4 treatment-emergent adverse events. Subgroup analyses by age, International Staging System stage, and cytogenetic risk were consistent with the overall population. KRd is an effective and tolerable treatment option for patients with NDMM regardless of HDM-ASCT eligibility.
Collapse
Affiliation(s)
- Ola Landgren
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | | | - Murielle Roussel
- Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Jagoda Jasielec
- Internal Medicine - Hematology, University of Chicago, Chicago, IL, USA
| | - Dominik Dytfeld
- Department of Hematology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | | |
Collapse
|