101
|
Palmiero G, Milani P, Verrillo F, Monda E, Merlo M, Emdin M, Limongelli G, Palladini G. Advances in the Therapy of Light Chain Cardiac Amyloidosis. Heart Fail Clin 2024; 20:e45-e54. [PMID: 40082015 DOI: 10.1016/j.hfc.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Systemic light chain (AL) amyloidosis stems from abnormal production of amyloidogenic immunoglobulin light chains by plasma cells or B-cell disorders. It can present locally or systemically, with systemic forms posing significant mortality risks, especially if cardiac involvement is present. Delayed diagnosis due to nonspecific symptoms leads to progressive organ dysfunction. Early recognition is vital for timely treatment, aiming to suppress amyloid production and reduce organ damage, thus promoting recovery and improving survival rates. This review article provides a comprehensive overview of the recent advancements in medical therapy for patients with AL cardiac amyloidosis.
Collapse
Affiliation(s)
- Giuseppe Palmiero
- Department of Translational Medical Sciences, Inherited and Rare Cardiovascular Diseases Unit, University of Campania "Luigi Vanvitelli", Naples, Italy; European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart-ERN GUARD Heart, Via P. Valdoni 7, Trieste 34100, Italy.
| | - Paolo Milani
- Amyloidosis Research and Treatment Center, Foundation "Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo", Pavia, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Federica Verrillo
- Department of Translational Medical Sciences, Inherited and Rare Cardiovascular Diseases Unit, University of Campania "Luigi Vanvitelli", Naples, Italy; European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart-ERN GUARD Heart, Via P. Valdoni 7, Trieste 34100, Italy
| | - Emanuele Monda
- Department of Translational Medical Sciences, Inherited and Rare Cardiovascular Diseases Unit, University of Campania "Luigi Vanvitelli", Naples, Italy; European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart-ERN GUARD Heart, Via P. Valdoni 7, Trieste 34100, Italy
| | - Marco Merlo
- European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart-ERN GUARD Heart, Via P. Valdoni 7, Trieste 34100, Italy; Cardiovascular Department, Azienda Sanitaria Universitaria Giuliano-Isontina (ASUGI) and University of Trieste, Trieste 34100, Italy
| | - Michele Emdin
- Scuola Superiore Sant'Anna, Pisa, Italy; Fondazione Toscana Gabriele Monasterio, via G. Moruzzi 1, Pisa 56124, Italy
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, Inherited and Rare Cardiovascular Diseases Unit, University of Campania "Luigi Vanvitelli", Naples, Italy; European Reference Network for Rare Low Prevalence and Complex Diseases of the Heart-ERN GUARD Heart, Via P. Valdoni 7, Trieste 34100, Italy; Institute of Cardiovascular Science, University College London, London, UK
| | - Giovanni Palladini
- Amyloidosis Research and Treatment Center, Foundation "Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo", Pavia, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
102
|
Christian LM, Kiecolt-Glaser JK, Cole SW, Burd CE, Madison AA, Wilson SJ, Rosko AE. Psychoneuroimmunology in multiple myeloma and autologous hematopoietic stem cell transplant: Opportunities for research among patients and caregivers. Brain Behav Immun 2024; 119:507-519. [PMID: 38643954 DOI: 10.1016/j.bbi.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024] Open
Abstract
Multiple myeloma (MM) is an incurable cancer and is the leading indication for autologous hematopoietic stem cell transplantation (HSCT). To be eligible for HSCT, a patient must have a caregiver, as caregivers play a central role in HSCT preparation and recovery. MM patients remain on treatment indefinitely, and thus patients and their caregivers face long-term challenges including the intensity of HSCT and perpetual therapy after transplant. Importantly, both patients and their caregivers show heightened depressive and anxiety symptoms, with dyadic correspondence evidenced and caregivers' distress often exceeding that of patients. An extensive psychoneuroimmunology (PNI) literature links distress with health via immune and neuroendocrine dysregulation as well as biological aging. However, data on PNI in the context of multiple myeloma - in patients or caregivers - are remarkably limited. Distress in MM patients has been associated with poorer outcomes including higher inflammation, greater one year post-HSCT hospital readmissions, and worse overall survival. Further, anxiety and depression are linked to biological aging and may contribute to the poor long-term health of both patients and caregivers. Because MM generally affects older adults, individual differences in biological aging may represent an important modifier of MM biology and HSCT treatment outcomes. There are a number of clinical scenarios in which biologically younger people could be prescribed more intensive therapies, with potential for greater benefit, by using a personalized cancer therapy approach based on the quantification of physiologic reserve. Further, despite considerable psychological demands, the effects of distress on health among MM caregivers is largely unexamined. Within this context, the current critical review highlights gaps in knowledge at the intersection of HSCT, inflammation, and biological aging in the context of MM. Research in this area hold promise for opportunities for novel and impactful psychoneuroimmunology (PNI) research to enhance health outcomes, quality of life, and longevity among both MM patients and their caregivers.
Collapse
Affiliation(s)
- Lisa M Christian
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA; The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Janice K Kiecolt-Glaser
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Steve W Cole
- Departments of Psychiatry and Biobehavioral Sciences and Medicine, Division of Hematology-Oncology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Christin E Burd
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA; Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Annelise A Madison
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, Columbus, OH 43210, USA; Veteran's Affairs Boston Healthcare System, Boston, MA 02130, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| | - Stephanie J Wilson
- Department of Psychology, Southern Methodist University, Dallas, TX 75206, USA
| | - Ashley E Rosko
- Division of Hematology, James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
103
|
Malard F, Neri P, Bahlis NJ, Terpos E, Moukalled N, Hungria VTM, Manier S, Mohty M. Multiple myeloma. Nat Rev Dis Primers 2024; 10:45. [PMID: 38937492 DOI: 10.1038/s41572-024-00529-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/16/2024] [Indexed: 06/29/2024]
Abstract
Multiple myeloma (MM) is a haematological lymphoid malignancy involving tumoural plasma cells and is usually characterized by the presence of a monoclonal immunoglobulin protein. MM is the second most common haematological malignancy, with an increasing global incidence. It remains incurable because most patients relapse or become refractory to treatments. MM is a genetically complex disease with high heterogeneity that develops as a multistep process, involving acquisition of genetic alterations in the tumour cells and changes in the bone marrow microenvironment. Symptomatic MM is diagnosed using the International Myeloma Working Group criteria as a bone marrow infiltration of ≥10% clonal plasma cells, and the presence of at least one myeloma-defining event, either standard CRAB features (hypercalcaemia, renal failure, anaemia and/or lytic bone lesions) or biomarkers of imminent organ damage. Younger and fit patients are considered eligible for transplant. They receive an induction, followed by consolidation with high-dose melphalan and autologous haematopoietic cell transplantation, and maintenance therapy. In older adults (ineligible for transplant), the combination of daratumumab, lenalidomide and dexamethasone is the preferred option. If relapse occurs and requires further therapy, the choice of therapy will be based on previous treatment and response and now includes immunotherapies, such as bi-specific monoclonal antibodies and chimeric antigen receptor T cell therapy.
Collapse
Affiliation(s)
- Florent Malard
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Alexandra General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nour Moukalled
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | | | - Salomon Manier
- Department of Hematology, Lille University Hospital and INSERM UMR-S1277 and CNRS UMR9020, Lille, France
| | - Mohamad Mohty
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| |
Collapse
|
104
|
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
|
105
|
Patel K, Ivanov A, Jocelyn T, Hantel A, Garcia JS, Abel GA. Patient-Reported Outcomes in Phase 3 Clinical Trials for Blood Cancers: A Systematic Review. JAMA Netw Open 2024; 7:e2414425. [PMID: 38829615 PMCID: PMC11148691 DOI: 10.1001/jamanetworkopen.2024.14425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 06/05/2024] Open
Abstract
Importance Published research suggests that patient-reported outcomes (PROs) are neither commonly collected nor reported in randomized clinical trials (RCTs) for solid tumors. Little is known about these practices in RCTs for hematological malignant neoplasms. Objective To evaluate the prevalence of PROs as prespecified end points in RCTs of hematological malignant neoplasms, and to assess reporting of PROs in associated trial publications. Evidence Review All issues of 8 journals known for publishing high-impact RCTs (NEJM, Lancet, Lancet Hematology, Lancet Oncology, Journal of Clinical Oncology, Blood, JAMA, and JAMA Oncology) between January 1, 2018, and December 13, 2022, were searched for primary publications of therapeutic phase 3 trials for adults with hematological malignant neoplasms. Studies that evaluated pretransplant conditioning regimens, graft-vs-host disease treatment, or radiotherapy as experimental treatment were excluded. Data regarding trial characteristics and PROs were extracted from manuscripts and trial protocols. Univariable analyses assessed associations between trial characteristics and PRO collection or reporting. Findings Ninety RCTs were eligible for analysis. PROs were an end point in 66 (73%) trials: in 1 trial (1%) as a primary end point, in 50 (56%) as a secondary end point, and in 15 (17%) as an exploratory end point. PRO data were reported in 26 of 66 primary publications (39%): outcomes were unchanged in 18 and improved in 8, with none reporting worse PROs with experimental treatment. Trials sponsored by for-profit entities were more likely to include PROs as an end point (49 of 55 [89%] vs 17 of 35 [49%]; P < .001) but were not significantly more likely to report PRO data (20 of 49 [41%] vs 6 of 17 [35%]; P = .69). Compared with trials involving lymphoma (18 of 29 [62%]) or leukemia or myelodysplastic syndrome (18 of 28 [64%]), those involving plasma cell disorders or multiple myeloma (27 of 30 [90%]) or myeloproliferative neoplasms (3 of 3 [100%]) were more likely to include PROs as an end point (P = .03). Similarly, compared with trials involving lymphoma (3 of 18 [17%]) or leukemia or myelodysplastic syndrome (5 of 18 [28%]), those involving plasma cell disorders or multiple myeloma (16 of 27 [59%]) or myeloproliferative neoplasms (2 of 3 [67%]) were more likely to report PROs in the primary publication (P = .01). Conclusions and Relevance In this systematic review, almost 3 of every 4 therapeutic RCTs for blood cancers collected PRO data; however, only 1 RCT included PROs as a primary end point. Moreover, most did not report resulting PRO data in the primary publication and when reported, PROs were either better or unchanged, raising concern for publication bias. This analysis suggests a critical gap in dissemination of data on the lived experiences of patients enrolled in RCTs for hematological malignant neoplasms.
Collapse
Affiliation(s)
- Kishan Patel
- Department of Internal Medicine, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Alexandra Ivanov
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tajmah Jocelyn
- Center for Clinical Investigation, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Andrew Hantel
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jacqueline S. Garcia
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gregory A. Abel
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
106
|
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
|
107
|
Raje NS, Cohen AD, Patel KK, van de Donk NWCJ, Richter J, San-Miguel J. Practical Aspects of Immunotherapy: A Report from the 20th International Myeloma Society (IMS) Annual Meeting. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:350-357. [PMID: 38627181 DOI: 10.1016/j.clml.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 03/06/2024] [Indexed: 06/01/2024]
Abstract
Immunotherapeutic strategies, specifically T-cell-redirected therapies, have been transformative in the context of multiple myeloma (MM). With the approval of two chimeric antigen receptor T-cell (CAR-T) drug products and three bispecific antibodies/T-cell engagers (bsAbs/TCEs) in relapsed/refractory MM (RRMM), the 20th annual IMS meeting dedicated a session to the practical aspects of these therapies. Here, we highlight the discussion during this session, including the role of CAR-T and bsAb therapies in frontline MM treatment, management of acute toxicities, prevention and management of infections, and finally treatment sequencing of T-cell redirected therapies.
Collapse
Affiliation(s)
- Noopur S Raje
- Division of Hematology/Oncology, Massachusetts General Hospital Cancer Center, Boston, MA.
| | - Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Krina K Patel
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Niels W C J van de Donk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Hematology, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Joshua Richter
- Tisch Cancer Institute Icahn School of Medicine at Mount Sinai New York, New York, NY
| | - Jesus San-Miguel
- Cancer Center Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| |
Collapse
|
108
|
Obajed Al-Ali N, Pinczes LI, Farkas K, Kerekes G, Illes A, Varoczy L. Steady-State Versus Chemotherapy-Based Stem Cell Mobilization in Multiple Myeloma: A Single-Center Study to Analyze Efficacy and Safety. J Hematol 2024; 13:79-85. [PMID: 38993736 PMCID: PMC11236354 DOI: 10.14740/jh1256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/29/2024] [Indexed: 07/13/2024] Open
Abstract
Background High-dose chemotherapy followed by autologous hematopoietic stem cell support is recommended in the treatment of eligible multiple myeloma (MM) patients. The aim of this study was to compare the efficacy and safety of steady-state versus chemotherapy-based stem cell mobilization in our Hungarian patient population. Methods The subjects were 210 MM patients who underwent stem cell mobilization procedure between 2018 and 2022. Solo granulocyte colony-stimulating factor (G-CSF) was administered in 104 cases, while 106 patients received chemotherapy which was followed by G-CSF administration. We evaluated the ratio of successful mobilizations, the amount of collected stem cells, the incidence of infections and cost-effectivity in the two groups. Results In the steady-state group, there was a significantly higher need for plerixafor (45% vs. 13%, P < 0.001), unsuccessful stem cell mobilization was more frequent (11% vs. 3%, P = 0.024) and the mean amount of collected stem cells was lower (6.9 vs. 9.8 × 106, P < 0.001) than in the chemotherapy group. However, infections were less frequent (4% vs. 27%, P < 0.001) and the number of days spent in hospital was significantly lower (6 vs. 14 days, P < 0.001). Plerixafor was more frequently administered in those who had received lenalidomide or daratumumab than in those who had been treated with other regimens (41% vs. 23%, P = 0.007 and 78% vs. 23%, P < 0.001, respectively). Conclusions Steady-state mobilization is a safe method; however, the higher rate of plerixafor administration and unsuccessful attempts may question its superiority to chemomobilization.
Collapse
Affiliation(s)
- Nora Obajed Al-Ali
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Sciences, University of Debrecen, Debrecen, Hungary
| | - Laszlo Imre Pinczes
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Sciences, University of Debrecen, Debrecen, Hungary
| | - Katalin Farkas
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Sciences, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Kerekes
- Intensive Care Unit, Institute for Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Arpad Illes
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Sciences, University of Debrecen, Debrecen, Hungary
| | - Laszlo Varoczy
- Department of Hematology, Institute for Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Clinical Sciences, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
109
|
Mori Y, Takizawa J, Katsuoka Y, Takezako N, Nagafuji K, Handa H, Kuroda J, Sunami K, Kamimura T, Ogawa R, Kikushige Y, Harada M, Akashi K, Miyamoto T, The Japan Study Group for Cell Therapy and Transplantation. Autologous HSCT with novel agent-based induction and consolidation followed by lenalidomide maintenance for untreated multiple myeloma. Cancer Sci 2024; 115:2002-2011. [PMID: 38498976 PMCID: PMC11145111 DOI: 10.1111/cas.16158] [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: 01/14/2024] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024] Open
Abstract
Triplet regimen comprising proteasome inhibitors, immunomodulatory drugs, and dexamethasone (DEX) is a recommended induction/consolidation therapy for multiple myeloma (MM) patients eligible for transplant. In this Japanese phase II study conducted from 2017 to 2019, newly diagnosed MM patients aged 20-65 received four induction cycles with bortezomib (Bor), lenalidomide (Len), and DEX (VRD), followed by Bor and high-dose melphalan with autologous stem cell rescue. Subsequently, they underwent four consolidation cycles with carfilzomib, Len, and DEX (KRD), followed by Len maintenance until disease progression. A total of 141 patients were analyzed. In an intent-to-treat population, the complete or better response post induction was 19.9%, rising to 39.7%, 58.9%, and 62.4% after transplant, consolidation, and 1-year maintenance, respectively. With a median follow-up of 38 months, the 3-year progression-free survival (PFS) rate was 83.5% and the 3-year overall survival rate was 92.5%. Severe adverse events (≥grade 3) occurred in ~30% of patients; however, there was no treatment-related mortality. These findings clearly showed the tolerability and effectiveness of this protocol. Nevertheless, patients with high-risk cytogenetics showed a trend toward lower 3-year PFS than those without (77.8% vs. 89.4%, p = 0.051), and ultra-high-risk cytogenetics (≥2 high-risk cytogenetics) had an even worse prognosis, with 61.2% 3-year PFS. To overcome this situation, a more potent treatment strategy incorporating novel agents such as the CD38-antibody should be assessed in future studies.
Collapse
Affiliation(s)
- Yasuo Mori
- Department of Medicine and Biosystemic SciencesKyushu University Graduate School of MedicineFukuokaJapan
| | - Jun Takizawa
- Division of Hematology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yuna Katsuoka
- Department of HematologyNational Hospital Organization Sendai Medical CenterSendaiJapan
| | - Naoki Takezako
- Department of HematologyNational Hospital Organization Disaster Medical CenterTokyoJapan
- Division of Hematology, Japan Association for Development of Community MedicineNerima Hikarigaoka HospitalTokyoJapan
| | - Koji Nagafuji
- Division of Hematology and Oncology, Department of MedicineKurume University School of MedicineKurumeJapan
| | - Hiroshi Handa
- Department of HematologyGunma University Graduate School of MedicineMaebashiGunmaJapan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Kazutaka Sunami
- Department of HematologyNational Hospital Organization Okayama Medical CenterOkayamaJapan
| | | | - Ryosuke Ogawa
- Department of Hematology and OncologyJCHO Kyushu HospitalFukuokaJapan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic SciencesKyushu University Graduate School of MedicineFukuokaJapan
| | - Mine Harada
- Karatsu Higashimatsuura Medical CenterKaratsuJapan
| | - Koichi Akashi
- Department of Medicine and Biosystemic SciencesKyushu University Graduate School of MedicineFukuokaJapan
| | - Toshihiro Miyamoto
- Department of Medicine and Biosystemic SciencesKyushu University Graduate School of MedicineFukuokaJapan
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health SciencesKanazawa UniversityKanazawaIshikawaJapan
| | | |
Collapse
|
110
|
Quivoron C, Michot JM, Danu A, Lecourt H, Saada V, Saleh K, Vergé V, Cotteret S, Bernard OA, Ribrag V. Sensitivity, specificity, and accuracy of molecular profiling on circulating cell-free DNA in refractory or relapsed multiple myeloma patients, results of MM-EP1 study. Leuk Lymphoma 2024; 65:789-799. [PMID: 38433500 DOI: 10.1080/10428194.2024.2320258] [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: 11/15/2023] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
As a promising alternative to bone marrow aspiration (BMA), mutational profiling on blood-derived circulating cell-free tumor DNA (cfDNA) is a harmless and simple technique to monitor molecular response and treatment resistance of patients with refractory/relapsed multiple myeloma (R/R MM). We evaluated the sensitivity and specificity of cfDNA compared to BMA CD138 positive myeloma plasma cells (PCs) in a series of 45 R/R MM patients using the 29-gene targeted panel (AmpliSeq) NGS. KRAS, NRAS, FAM46C, DIS3, and TP53 were the most frequently mutated genes. The average sensitivity and specificity of cfDNA detection were 65% and 97%, respectively. The concordance per gene between the two samples was good to excellent according to Cohen's κ coefficients interpretation. An increased number of mutations detected in cfDNA were associated with a decreased overall survival. In conclusion, we demonstrated cfDNA NGS analysis feasibility and accuracy in R/R MM patients who may benefit from early phase clinical trial.
Collapse
Affiliation(s)
- C Quivoron
- Translational Hematology Laboratory, AMMICa, INSERM US23/CNRS UAR3655, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - J-M Michot
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
- Drug Development Department: Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - A Danu
- Hematology Department, Gustave Roussy, Villejuif, France
| | - H Lecourt
- Translational Hematology Laboratory, AMMICa, INSERM US23/CNRS UAR3655, Gustave Roussy Cancer Campus, Villejuif, France
| | - V Saada
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - K Saleh
- Hematology Department, Gustave Roussy, Villejuif, France
| | - V Vergé
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - S Cotteret
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - O A Bernard
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - V Ribrag
- Translational Hematology Laboratory, AMMICa, INSERM US23/CNRS UAR3655, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
- Drug Development Department: Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
- Hematology Department, Gustave Roussy, Villejuif, France
| |
Collapse
|
111
|
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
|
112
|
Shibayama H, Itagaki M, Handa H, Yokoyama A, Saito A, Kosugi S, Ota S, Yoshimitsu M, Tanaka Y, Kurahashi S, Fuchida SI, Iino M, Shimizu T, Moriuchi Y, Toyama K, Mitani K, Tsukune Y, Kada A, Tamura H, Abe M, Iwasaki H, Kuroda J, Takamatsu H, Sunami K, Kizaki M, Ishida T, Saito T, Matsumura I, Akashi K, Iida S. Primary analysis of a prospective cohort study of Japanese patients with plasma cell neoplasms in the novel drug era (2016-2021). Int J Hematol 2024; 119:707-721. [PMID: 38548963 PMCID: PMC11136844 DOI: 10.1007/s12185-024-03754-8] [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: 09/13/2023] [Revised: 03/04/2024] [Accepted: 03/08/2024] [Indexed: 05/31/2024]
Abstract
The emergence of novel drugs has significantly improved outcomes of patients with plasma cell neoplasms (PCN). The Japanese Society of Hematology conducted a prospective observational study in newly diagnosed PCN patients between 2016 and 2021. The analysis focused on 1385 patients diagnosed with symptomatic PCN between 2016 and 2018. The primary endpoint was the 3-year overall survival (OS) rate among patients requiring treatment (n = 1284), which was 70.0% (95%CI 67.4-72.6%). Approximately 94% of these patients received novel drugs as frontline therapy. The 3-year OS rate was 90.3% (95%CI 86.6-93.1%) in the 25% of patients who received upfront autologous stem cell transplantation (ASCT), versus just 61.4% (95%CI 58.0-64.6%) in those who did not receive upfront ASCT. The only unfavorable prognostic factor that affected OS in ASCT recipients was an age of 65 or higher. For patients who did not receive ASCT, independent unfavorable prognostic factors included frontline treatment with conventional chemotherapies, international staging system score of 2/3, extramedullary tumors, and Freiberg comorbidity index of 2/3. This study unequivocally demonstrates that use of novel drugs improved OS in Japanese myeloma patients, and underscores the continued importance of upfront ASCT as the standard of care in the era of novel drugs.
Collapse
Affiliation(s)
- Hirohiko Shibayama
- Department of Hematology, NHO Osaka National Hospital, 2-1-14 Hoenzaka Chuo-ku, Osaka City, Osaka, 540-0006, Japan.
- Department of Hematology and Oncology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Mitsuhiro Itagaki
- Department of Hematology, Hiroshima Red Cross Hospital, Hiroshima, Japan
| | - Hiroshi Handa
- Department of Hematology, Gunma University, Gunma, Japan
| | - Akihiro Yokoyama
- Department of Hematology, NHO Tokyo Medical Center, Tokyo, Japan
| | - Akio Saito
- Department of Hematology, NHO Shibukawa Medical Center, Gunma, Japan
| | - Satoru Kosugi
- Department of Hematology, Toyonaka Municipal Hospital, Osaka, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Hokkaido, Japan
| | - Makoto Yoshimitsu
- Department of Hematology and Rheumatology, Kagoshima University, Kagoshima, Japan
| | | | - Shingo Kurahashi
- Department of Hematology and Oncology, Toyohashi Municipal Hospital, Aichi, Japan
| | - Shin-Ichi Fuchida
- Department of Hematology, JCHO Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Masaki Iino
- Department of Hematology, Yamanashi Prefectural Central Hospital, Yamanashi, Japan
| | - Takayuki Shimizu
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | - Kohtaro Toyama
- Department of Hematology, Fujioka General Hospital, Gunma, Japan
| | - Kinuko Mitani
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| | - Yutaka Tsukune
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akiko Kada
- Clinical Research Center, NHO Nagoya Medical Center, Aichi, Japan
| | - Hideto Tamura
- Division of Diabetes, Endocrinology and Hematology, Department of Internal Medicine, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Masahiro Abe
- Department of Hematology, Kawashima Hospital, Tokushima, Japan
| | - Hiromi Iwasaki
- Department of Hematology, NHO Kyushu Medical Center, Fukuoka, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Department of Medicine, Kyoto Prefecture University of Medicine, Kyoto, Japan
| | | | - Kazutaka Sunami
- Department of Hematology, NHO Okayama Medical Center, Okayama, Japan
| | - Masahiro Kizaki
- Department of Hematology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Tadao Ishida
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Toshiki Saito
- Clinical Research Center, NHO Nagoya Medical Center, Aichi, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University, Osaka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University, Fukuoka, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Institute of Medical and Pharmaceutical Sciences, 1 Kawasumi, Mizuho-Cho, Mizuho-ku, Nagoya City, Aichi, 467-8601, Japan.
| |
Collapse
|
113
|
Nooka AK, Kaufman JL, Rodriguez C, Jakubowiak A, Efebera Y, Reeves B, Wildes TM, Holstein SA, Anderson LD, Badros A, Shune L, Chari A, Pei H, Cortoos A, Patel S, Lin TS, Voorhees PM, Usmani SZ, Richardson PG. Post hoc analysis of daratumumab plus lenalidomide, bortezomib and dexamethasone in Black patients from final data of the GRIFFIN study. Br J Haematol 2024; 204:2227-2232. [PMID: 38504552 DOI: 10.1111/bjh.19386] [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: 10/13/2023] [Revised: 02/14/2024] [Accepted: 02/24/2024] [Indexed: 03/21/2024]
Abstract
Due in part to racial disparities and underrepresentation in clinical studies, optimal therapies for Black patients with multiple myeloma remain undefined. This final analysis of GRIFFIN by race showed that the addition of daratumumab (D) to lenalidomide/bortezomib/dexamethasone (RVd) provides clinical benefit among both Black and White transplant-eligible newly diagnosed patients compared with RVd alone. However, Black patients were more likely to discontinue ≥1 drug due to treatment-emergent adverse events. In summary, these findings suggest a benefit of D-RVd front-line therapy among Black and White patients and underscore the importance of equitable treatment access for all patients.
Collapse
Affiliation(s)
- Ajay K Nooka
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | | | - Cesar Rodriguez
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Brandi Reeves
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tanya M Wildes
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sarah A Holstein
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Larry D Anderson
- Myeloma, Waldenström's and Amyloidosis Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ashraf Badros
- Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland, USA
| | - Leyla Shune
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ajai Chari
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
- University of California at San Francisco, San Francisco, California, USA
| | - Huiling Pei
- Janssen Research & Development, LLC, Titusville, New Jersey, USA
| | | | - Sharmila Patel
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, USA
| | - Thomas S Lin
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, USA
| | - Peter M Voorhees
- Levine Cancer Institute, Atrium Health Wake Forest University School of Medicine, Charlotte, North Carolina, USA
| | - Saad Z Usmani
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
114
|
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
|
115
|
Rees MJ, D'Agostino M, Leypoldt LB, Kumar S, Weisel KC, Gay F. Navigating High-Risk and Ultrahigh-Risk Multiple Myeloma: Challenges and Emerging Strategies. Am Soc Clin Oncol Educ Book 2024; 44:e433520. [PMID: 38772002 DOI: 10.1200/edbk_433520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Despite significant improvement in the outcomes of patients with newly diagnosed multiple myeloma (NDMM) with novel therapies, there is still an underserved high-risk (HR) population that experiences early disease progression and death. With the median survival crossing 10 years, we defined ultrahigh-risk (uHR)MM as MM leading to death within 24-36 months of diagnosis and HRMM as MM leading to death within 36-60 months. Several features have emerged as markers of uHRMM: the co-occurrence of two or more high-risk cytogenetic abnormalities, extramedullary disease, plasma cell leukemia and a high-risk gene expression profiling signature. The heterogeneous risk definition across trials, the few trials available designed for HR patients, and the small HR subgroups in all-comers trials make it difficult to generate recommendations with high levels of evidence. Nevertheless, regardless of treatment administered, several studies consistently showed that achieving and maintaining measurable residual disease negativity is now considered the main factor able to mitigate the adverse prognosis related to baseline features. For fit patients with HR transplant-eligible (TE) NDMM, quadruplet induction/consolidation treatment with anti-CD38 monoclonal antibodies, immunomodulatory agents, proteasome inhibitors and dexamethasone, and autologous stem-cell transplant and maintenance with, if available, at least a doublet combination could be considered the option of choice. For non-TE NDMM, considering the recent data generated and carefully reviewing those upcoming, quadruplet treatment consisting of anti-CD38 monoclonal antibodies, immunomodulatory agents, proteasome inhibitors, and dexamethasone should also be considered. Future trials integrating BCMA-directed novel generation immunotherapies hold great potential for further advancing the treatment landscape in all NDMM patients with HR disease.
Collapse
Affiliation(s)
| | - Mattia D'Agostino
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, AOU Città della Salute e della Scienza, University of Torino, Torino, Italy
| | - Lisa B Leypoldt
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Katja C Weisel
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Gay
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, AOU Città della Salute e della Scienza, University of Torino, Torino, Italy
| |
Collapse
|
116
|
Fazio F, Passucci M, Micozzi J, Di Landro F, Fianchi L, Za T, Manieri VM, Annibali O, Cupelli L, Bongarzoni V, Gentili S, De Padua L, Crisanti E, Garzia MG, Rago A, Piciocchi A, Mengarelli A, Morè S, De Stefano V, Bafti MS, Martelli M, Petrucci MT. Effect of daratumumab on stem cell yields in patients with newly diagnosed multiple myeloma: a report from the Multiple Myeloma Group. Bone Marrow Transplant 2024; 59:896-898. [PMID: 38461292 PMCID: PMC11161397 DOI: 10.1038/s41409-024-02260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024]
Affiliation(s)
- F Fazio
- Hematology, Azienda Policlinico Umberto I-Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| | - M Passucci
- Hematology, Azienda Policlinico Umberto I-Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - J Micozzi
- Hematology, Azienda Policlinico Umberto I-Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - F Di Landro
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - L Fianchi
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - T Za
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - V M Manieri
- Clinica di Ematologia Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - O Annibali
- Unit of Hematology, Stem Cell Transplantation, University Campus Bio-Medico, Rome, Italy
| | - L Cupelli
- UOC Hematology, Hospital S. Eugenio, Rome, Italy
| | - V Bongarzoni
- Department of Hematology San Giovanni-Addolorata Hospital, Rome, Italy
| | - S Gentili
- UOSD Ematologia Civitanova Marche AST Macerata, Rome, Italy
| | - L De Padua
- Hematology Unit, Fabrizio Spaziani Hospital, Frosinone, Italy
| | - E Crisanti
- Hematology and Transplant Unit, Santa Maria Goretti Hospital, AUSL, Latina, Italy
| | - M G Garzia
- Department of Hematology, Hematology San Camillo Forlanini Hospital Rome Italy, Rome, Italy
| | - A Rago
- UOSD Ematologia ASL Roma1, Rome, Italy
| | - A Piciocchi
- Italian Group for Adult Hematologic Diseases (GIMEMA) Data Center, Rome, Italy
| | - A Mengarelli
- Hematology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - S Morè
- Clinica di Ematologia Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - V De Stefano
- Section of Hematology, Department of Radiological and Hematological Sciences, Catholic University, Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - M S Bafti
- Department of Immuno-Hematology and Transfusional Medicine, Policlinico Umberto I, Rome, Italy
| | - M Martelli
- Hematology, Azienda Policlinico Umberto I-Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - M T Petrucci
- Hematology, Azienda Policlinico Umberto I-Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
117
|
Buck T, Hartley-Brown MA, Efebera YA, Milner CP, Zonder JA, Richardson PG, Salinardi T, Rice MS. Real-world multiple myeloma risk factors and outcomes by non-Hispanic Black/African American and non- Hispanic White race/ethnicity in the United States. Haematologica 2024; 109:1882-1892. [PMID: 38031762 PMCID: PMC11141648 DOI: 10.3324/haematol.2023.282788] [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: 01/25/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Examination of the impact of race and ethnicity on multiple myeloma (MM) outcomes has yielded inconsistent results. This retrospective, real-world (RW) study describes patient, disease, and treatment characteristics (and associations with survival outcomes) among newly diagnosed MM patients of non-Hispanic (NH) Black/African American (AA) and NH White race/ethnicity in the US. We included patients from the nationwide Flatiron Health electronic health record-derived de-identified database who initiated first line of therapy (LOT) for MM between January 1, 2016 and March 31, 2022. Of 4,614 patients in our study cohort, 23.3% were NH Black/AA. Non-Hispanic Black/AA patients were younger than NH White patients at diagnosis (median 68 vs. 71 years) and more likely to be female (53.4% vs. 43.5%). Rates of high-risk cytogenetics and 1q21+ were similar between races/ethnicities. The most common primary regimen used was lenalidomide-bortezomib-dexamethasone (50.1% of NH Black/AA and 48.1% of NH White patients). Receipt of stem cell transplantation during first LOT was less common among NH Black/AA (16.5%) than NH White (21.9%) patients. Unadjusted RW progression-free survival (rwPFS) and overall survival (rwOS) were similar between races/ethnicities. After multivariable adjustment, NH Black/AA race/ethnicity was associated with slightly inferior rwPFS (hazard ratio [HR]=1.13; 95% confidence interval [CI]: 1.01-1.27). The difference in rwOS (HR=1.12; 95% CI: 0.98-1.28) was not statistically significant. In general, associations between risk factors for rwPFS and rwOS were consistent between races/ethnicities. Findings from this analysis help to inform clinicians about the impact of race/ethnicity on MM treatment paradigms and outcomes in the US.
Collapse
Affiliation(s)
- Tondre Buck
- Spartanburg Medical Center, Center for Hematology/Oncology, Spartanburg, SC.
| | - Monique A Hartley-Brown
- Division of Hematologic Malignancy, Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA
| | - Yvonne A Efebera
- Division of Blood and Marrow Transplant and Cellular Therapy, OhioHealth, Columbus, OH
| | - Carter P Milner
- Division of Hematology and Medical Oncology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Jeffrey A Zonder
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Paul G Richardson
- Division of Hematologic Malignancy, Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA
| | | | | |
Collapse
|
118
|
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
|
119
|
Pasvolsky O, Wang Z, Milton DR, Tanner MR, Bashir Q, Srour S, Saini N, Lin P, Ramdial J, Nieto Y, Tang G, Kebriaei P, Aljawai Y, Khan HN, Lee HC, Ye C, Patel KK, Thomas SK, Orlowski RZ, Shpall EJ, Champlin RE, Qazilbash MH. Multiple myeloma patients with a long remission after autologous hematopoietic stem cell transplantation. Blood Cancer J 2024; 14:82. [PMID: 38760362 PMCID: PMC11101444 DOI: 10.1038/s41408-024-01062-2] [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] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
Autologous stem cell transplantation (autoHCT) is considered standard of care for newly diagnosed multiple myeloma (MM). Although most patients eventually progress after autoHCT, a small proportion achieve a durable response. In this retrospective study we included 1576 patients, 244 (15%) of whom were long-term responders (LTR), defined as having a progression-free survival (PFS) of ≥8 years after transplant. Patients in the LTR group were younger than the non-LTR group (median age 58.4 vs. 59.5 years; p = 0.012), less likely to have high-risk cytogenetics (4% vs. 14%; p < 0.001), more often had <50% bone marrow plasma cells (67% vs. 58%; p = 0.018) and R-ISS stage I disease (43% vs. 34%). More patients in the LTR group received post-transplant maintenance (63% vs. 52%; p = 0.002). Patients in the LTR group had higher rates of complete response (CR) at day100 (41% vs. 27%; p < 0.001) and at best post-transplant response (70% vs. 37%; p < 0.001), compared to the non-LTR group. Patients in the LTR groups had a median PFS of 169.3 months and the median overall survival (OS) had not been reached. The leading cause of death in the LTR was disease progression. In conclusion, 15% of patients in the cohort were LTR after upfront autoHCT, with distinct characteristics and a median PFS of more than 14 years.
Collapse
Affiliation(s)
- Oren Pasvolsky
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhongya Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Denái R Milton
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark R Tanner
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeraj Saini
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yosra Aljawai
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hina N Khan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematology/Oncology, McGovern Medical School, The University of Texas, Health Sciences Center at Houston, Houston, TX, USA
| | - Hans C Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine Ye
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Krina K Patel
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheeba K Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard E Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
120
|
[Chinese expert consensus on diagnosis and treatment of high risk multiple myeloma (2024)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:430-435. [PMID: 38964916 PMCID: PMC11270502 DOI: 10.3760/cma.j.cn121090-20240312-00090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Indexed: 07/06/2024]
Abstract
High-risk multiple myeloma (HRMM) refers to patients with multiple myeloma whose overall survival time is less than 2-3 years under current standardized diagnosis and treatment. By combining various static and dynamic prognostic factors, risk stratification is performed to identify HRMM patients early and treat patients with personalized strategies, with the aim of significantly improving adverse survival outcomes in HRMM patients. Although the clinical value of HRMM has reached a consensus domestically in recent years, there still exist confusions and ambiguities in the definition, high-risk factors, risk stratification, and treatment of HRMM, necessitating standardization. In order to enhance the diagnostic and treatment capabilities of Chinese physicians in HRMM, the Professional Committee of Hematologic Malignancies of the Chinese Anti-Cancer Association (CACA) and the Multiple Myeloma Expert Committee of the Chinese Society of Clinical Oncology (CSCO) have organized relevant experts to develop this consensus. This consensus aims to clarify the definition of HRMM, high-risk factors, and risk stratification system, and provide treatment recommendations for HRMM, thereby improving the quality of life and prognosis of Chinese HRMM patients.
Collapse
|
121
|
Mehl J, Akhoundova D, Bacher U, Jeker B, Rhyner Agocs G, Ruefer A, Soltermann S, Soekler M, Winkler A, Daskalakis M, Pabst T. Daratumumab during Myeloma Induction Therapy Is Associated with Impaired Stem Cell Mobilization and Prolonged Post-Transplant Hematologic Recovery. Cancers (Basel) 2024; 16:1854. [PMID: 38791933 PMCID: PMC11119719 DOI: 10.3390/cancers16101854] [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: 04/27/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
Daratumumab is being increasingly integrated into first-line multiple myeloma (MM) induction regimens, leading to improved response depth and longer progression-free survival. Autologous stem cell transplantation (ASCT) is commonly performed as a consolidation strategy following first-line induction in fit MM patients. We investigated a cohort of 155 MM patients who received ASCT after first-line induction with or without daratumumab (RVd, n = 110; D-RVd, n = 45), analyzing differences in stem cell mobilization, apheresis, and engraftment. In the D-RVd group, fewer patients successfully completed mobilization at the planned apheresis date (44% vs. 71%, p = 0.0029), and more patients required the use of rescue plerixafor (38% vs. 28%, p = 0.3052). The median count of peripheral CD34+ cells at apheresis was lower (41.37 vs. 52.19 × 106/L, p = 0.0233), and the total number of collected CD34+ cells was inferior (8.27 vs. 10.22 × 106/kg BW, p = 0.0139). The time to recovery of neutrophils and platelets was prolonged (12 vs. 11 days, p = 0.0164; and 16 vs. 14 days, p = 0.0002, respectively), and a higher frequency of erythrocyte transfusions (74% vs. 51%, p = 0.0103) and a higher number of platelet concentrates/patients were required (4 vs. 2; p = 0.001). The use of daratumumab during MM induction might negatively impact stem cell mobilization and engraftment in the context of ASCT.
Collapse
Affiliation(s)
- Julian Mehl
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (J.M.); (D.A.); (B.J.)
| | - Dilara Akhoundova
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (J.M.); (D.A.); (B.J.)
| | - Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (U.B.); (M.D.)
| | - Barbara Jeker
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (J.M.); (D.A.); (B.J.)
| | - Gaëlle Rhyner Agocs
- Department of Medical Oncology, HFR Fribourg-Hôpital Cantonal, 1708 Fribourg, Switzerland;
| | - Axel Ruefer
- Department of Hematology, Cantonal Hospital Lucerne, 6000 Lucerne, Switzerland;
| | - Susanne Soltermann
- Department of Oncology and Hematology, Bürgerspital Solothurn, 4500 Solothurn, Switzerland;
| | - Martin Soekler
- Department of Oncology and Hematology, Hospital Thun, 3600 Thun, Switzerland;
| | - Annette Winkler
- Department of Oncology and Hematology, Biel Hospital Center, 2501 Biel, Switzerland;
| | - Michael Daskalakis
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (U.B.); (M.D.)
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (J.M.); (D.A.); (B.J.)
| |
Collapse
|
122
|
Barberio J, Lash TL, Nooka AK, Naimi AI, Patzer RE, Kim C. Real-World Risk of Severe Cytopenias in Multiple Myeloma Patients Sequentially Treated with Immunomodulatory Drugs. Acta Haematol 2024; 148:135-147. [PMID: 38735288 DOI: 10.1159/000539127] [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: 01/30/2024] [Accepted: 04/19/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION Most multiple myeloma (MM) patients experience cytopenias, likely driven by both disease and treatment-related factors. Immunomodulatory agents (IMiDs), which form the backbone of most anti-myeloma regimens, are known to cause higher grade cytopenias. In this context, the impact of sequential IMiD treatments on cytopenia risk is unknown. METHODS We evaluated the cumulative risks of severe cytopenias following second line of therapy (LOT) initiation in 5,573 MM patients in the Flatiron Health database. Patients for whom both LOTs 1 and 2 contained IMiDs were considered "sequentially exposed"; those for whom neither contained IMiDs were "never exposed." RESULTS For the neutropenia outcome, compared to the never exposed, the sequentially exposed had the highest 1-year risk (risk difference [RD] 12%), followed by those only recently exposed during LOT 2 (RD 8%), then by those with only past exposure during LOT 1 (RD 5%). A similar pattern was observed for leukopenia, but no meaningful differences were observed for anemia or thrombocytopenia. The associations between sequential exposure, versus never, with neutropenia and leukopenia were even stronger among those with a recent cytopenia history. CONCLUSION Results suggest that sequential exposure to IMiDs is a risk factor for higher grade cytopenias. These findings have profound clinical implications in choosing newer LOTs with potential risks of cytopenia.
Collapse
Affiliation(s)
- Julie Barberio
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| | - Timothy L Lash
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Ajay K Nooka
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ashley I Naimi
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Rachel E Patzer
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
- Regenstrief Institute, Indianapolis, Indiana, USA
| | - Christopher Kim
- Center for Observational Research, Amgen Inc., Thousand Oaks, California, USA
| |
Collapse
|
123
|
Cohen I, Vaxman I, Gertz MA. Historical Perspective of High-Dose Therapy Followed by Autologous Stem Cell Transplantation in Multiple Myeloma. Acta Haematol 2024; 148:289-299. [PMID: 38710160 DOI: 10.1159/000539225] [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: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND High-dose therapy (HDT) followed by autologous stem cell transplantation (ASCT) has become part of standard of care (SOC) in newly diagnosed multiple myeloma. In this review, we provide a historical perspective on ASCT since its introduction in the 1990s. SUMMARY Overall survival (OS) benefit for HDT followed by ASCT was demonstrated in studies comparing HDT with ASCT to standard-dose therapy (SDT) before the era of novel agents. Conditioning is done with melphalan 200 mg/m2. Lower doses (MEL140, MEL150) for older patients with comorbidities are safe and have comparable results. The addition of busulfan to melphalan improves progression-free survival (PFS) but not OS. HDT with ASCT after induction with novel agents prolongs PFS but not OS compared to SDT alone. The benefit is more evident in patients with high-risk cytogenetics. Mobilization can be achieved with granulocyte colony-stimulating factor alone, but is improved with the addition of chemotherapy. Plerixafor reduces mobilization failure and enables sufficient stem cell collection after induction with novel agents. ASCT is safe with a low rate of mortality (1%), and selected patients can be managed as outpatients. KEY MESSAGES HDT followed by ASCT remains part of SOC due to its PFS benefit and relatively low toxicity.
Collapse
Affiliation(s)
- Inbar Cohen
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel,
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel,
| | - Iuliana Vaxman
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
124
|
Saygin C, Zhang P, Stauber J, Aldoss I, Sperling AS, Weeks LD, Luskin MR, Knepper TC, Wanjari P, Wang P, Lager AM, Fitzpatrick C, Segal JP, Gharghabi M, Gurbuxani S, Venkataraman G, Cheng JX, Eisfelder BJ, Bohorquez O, Patel AA, Umesh Nagalakshmi S, Jayaram S, Odenike OM, Larson RA, Godley LA, Arber DA, Gibson CJ, Munshi NC, Marcucci G, Ebert BL, Greally JM, Steidl U, Lapalombella R, Shah BD, Stock W. Acute Lymphoblastic Leukemia with Myeloid Mutations Is a High-Risk Disease Associated with Clonal Hematopoiesis. Blood Cancer Discov 2024; 5:164-179. [PMID: 38150184 PMCID: PMC11061587 DOI: 10.1158/2643-3230.bcd-23-0106] [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: 06/23/2023] [Revised: 10/05/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023] Open
Abstract
Myeloid neoplasms arise from preexisting clonal hematopoiesis (CH); however, the role of CH in the pathogenesis of acute lymphoblastic leukemia (ALL) is unknown. We found that 18% of adult ALL cases harbored TP53, and 16% had myeloid CH-associated gene mutations. ALL with myeloid mutations (MyM) had distinct genetic and clinical characteristics, associated with inferior survival. By using single-cell proteogenomic analysis, we demonstrated that myeloid mutations were present years before the diagnosis of ALL, and a subset of these clones expanded over time to manifest as dominant clones in ALL. Single-cell RNA sequencing revealed upregulation of genes associated with cell survival and resistance to apoptosis in B-ALL with MyM, which responds better to newer immunotherapeutic approaches. These findings define ALL with MyM as a high-risk disease that can arise from antecedent CH and offer new mechanistic insights to develop better therapeutic and preventative strategies. SIGNIFICANCE CH is a precursor lesion for lymphoblastic leukemogenesis. ALL with MyM has distinct genetic and clinical characteristics, associated with adverse survival outcomes after chemotherapy. CH can precede ALL years before diagnosis, and ALL with MyM is enriched with activated T cells that respond to immunotherapies such as blinatumomab. See related commentary by Iacobucci, p. 142.
Collapse
Affiliation(s)
- Caner Saygin
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Pu Zhang
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | - Jacob Stauber
- Albert Einstein College of Medicine–Montefiore Health System, New York, New York
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | - Adam S. Sperling
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Hematology, Brigham and Women's Hospital, Boston, Massachusetts
| | | | | | | | - Pankhuri Wanjari
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Peng Wang
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Angela M. Lager
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | | - Jeremy P. Segal
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Mehdi Gharghabi
- Division of Hematology, The Ohio State University, Columbus, Ohio
| | | | | | - Jason X. Cheng
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Bart J. Eisfelder
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Oliver Bohorquez
- Albert Einstein College of Medicine–Montefiore Health System, New York, New York
| | - Anand A. Patel
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | | | | | | | - Richard A. Larson
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Lucy A. Godley
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| | - Daniel A. Arber
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | | | | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California
| | | | - John M. Greally
- Albert Einstein College of Medicine–Montefiore Health System, New York, New York
| | - Ulrich Steidl
- Albert Einstein College of Medicine–Montefiore Health System, New York, New York
| | | | | | - Wendy Stock
- Section of Hematology/Oncology, University of Chicago, Chicago, Illinois
| |
Collapse
|
125
|
Jagannath S, Jackson CC, Schecter JM, Lendvai N, Sun H, Akram M, Patel N, Martin TG. Cilta-cel, a BCMA-targeting CAR-T therapy for patients with multiple myeloma. Expert Opin Biol Ther 2024; 24:339-350. [PMID: 38738379 DOI: 10.1080/14712598.2024.2352591] [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: 01/04/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION Ciltacabtagene autoleucel (cilta-cel), a BCMA-targeting CAR-T therapy, is approved in the United States and Europe for patients with relapsed/refractory multiple myeloma (RRMM) and ≥1 prior line of therapy (LOT), including a proteasome inhibitor and an immunomodulatory drug, and are lenalidomide refractory. AREAS COVERED We examine recent long-term data in heavily pretreated RRMM (LEGEND-2, CARTITUDE-1) and earlier LOTs (CARTITUDE-4) compared with standard therapy and discuss the rationale for investigating cilta-cel as frontline therapy for transplant-eligible and transplant-ineligible patients (CARTITUDE-5, CARTITUDE-6). EXPERT OPINION CAR-T therapies can improve outcomes for patients with MM across different LOTs. CARTITUDE-1 and CARTITUDE-4 have set a new bar for efficacy, with median PFS of 34.9 months in heavily pretreated patients (CARTITUDE-1) and a 74% relative risk reduction for progression/death versus standard care in patients with 1-3 prior LOTs (CARTITUDE-4), with manageable safety. Response rates were consistent between the two studies: 98% in CARTITUDE-1 and approaching 100% for infused patients in CARTITUDE-4. Cilta-cel could be a key treatment choice for patients with RRMM after first LOT. Clinical trials investigating frontline cilta-cel therapy will provide valuable insights into optimizing treatment pathways with the aim to potentially cure MM.
Collapse
Affiliation(s)
- Sundar Jagannath
- Multiple Myeloma Division, Tisch Cancer Institute, Mount Sinai Medical Center New York, New York, NY, USA
| | | | | | | | - Huabin Sun
- Janssen Research & Development, Raritan, NJ, USA
| | | | | | - Thomas G Martin
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
126
|
Jurgens E, Usmani SZ. SOHO State of the Art Updates and Next Questions: Will CAR-T Replace ASCT in NDMM. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:277-284. [PMID: 38331676 DOI: 10.1016/j.clml.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 02/10/2024]
Abstract
The treatment landscape for multiple myeloma (MM) has rapidly evolved over the last 2 decades. The development of triplet and quadruplet regimens including proteasome inhibitors (PI), immunomodulatory agents (IMiDs), and anti-CD38 monoclonal antibodies has dramatically extended overall survival. In addition to effective multidrug regimens, autologous stem cell transplant (ASCT) is a cornerstone of management in newly diagnosed multiple myeloma (NDMM). However, despite these combined treatment modalities, curative therapy for MM remains elusive. Recent, novel immunotherapies including chimeric antigen T-cell (CAR-T) therapy have demonstrated deep and durable responses in relapsed and refractory multiple myeloma (RRMM). Currently 2 CAR-T products, ciltacabtagene autoleucel (cilta-cel) and idecabtagene vicleucel (ide-cel), are approved by the FDA for the treatment of RRMM. The success of CAR-T therapy revolutionized the management of RRMM prompting clinical trials studying CAR-T therapy in the first line setting. The ongoing KarMMa-4, CARTITUDE-5, and CARTITUDE-6 clinical trials may establish CAR-T therapy as a first line option potentially supplanting ASCT in the initial treatment of NDMM. In this review, we discuss the current standard of care management of NDMM, trace the evolution of CAR-T clinical trials in RRMM, and survey ongoing clinical trials studying CAR-T therapy in NDMM.
Collapse
Affiliation(s)
- Eric Jurgens
- Department of Medicine, Hematology Oncology Fellowship Program, MSKCC, New York, NY
| | - Saad Z Usmani
- Department of Medicine, Myeloma Service, MSKCC, New York, NY.
| |
Collapse
|
127
|
Wei JX, Shastri A, Sica RA, Mantzaris I, Kornblum N, Shah U, Janakiram M, Gritsman K, Verma A, Goldfinger M, Cooper D, Shah N. Impact of race and ethnicity on early mortality in multiple myeloma: a SEER analysis. Haematologica 2024; 109:1480-1486. [PMID: 37881838 PMCID: PMC11063841 DOI: 10.3324/haematol.2023.283304] [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: 04/12/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023] Open
Abstract
Over the past two decades, there have been significant advances in the treatment of multiple myeloma which has led to an improvement in overall survival.1,2 However, a notable proportion of patients continue to experience early mortality (EM), defined as 2 years from the time of diagnosis. This raises the possibility that improvements in myeloma survival have not extended equally to all groups. Using the latest data drawn from the Surveillance Epidemiology and End Results database of patients in the United States spanning 2000-2019, we study impact of important sociodemographic factors on EM. Through regression modeling, we demonstrate that patients diagnosed from 2000-2005, of older age, male sex, and of certain racial minority status (non-Hispanic Black and Hispanic) have higher odds of EM. Of these factors, minority status contributed to worse 2-year overall survival as well. We evaluate whether income, as a surrogate to access to care, could potentially explain this finding, but find that race has a distinct relationship with EM that is not modified by income. This is further reinforced by subgroup analysis. After characterizing groups vulnerable to EM, we examine reasons for these disparities and potential avenues to address them.
Collapse
Affiliation(s)
- John X Wei
- Department of Medicine, Montefiore/Albert Einstein College of Medicine, New York
| | - Aditi Shastri
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - R Alejandro Sica
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Ioannis Mantzaris
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Noah Kornblum
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Urvi Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - Murali Janakiram
- Division of Hematologic Malignancies, City of Hope National Medical Center, Duarte, CA
| | - Kira Gritsman
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Amit Verma
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Mendel Goldfinger
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Dennis Cooper
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York
| | - Nishi Shah
- Division of Hematologic Malignancies, Department of Medical Oncology, Montefiore/Albert Einstein College of Medicine, New York.
| |
Collapse
|
128
|
Tang HKK, Fung CY, Hwang YY, Lee H, Lau G, Yip SF, Kho B, Lau CK, Leung KH, Au E, Tse E, Sim J, Kwong YL, Chim CS. Prognostic factors in 448 newly diagnosed multiple myeloma receiving bortezomib-based induction: impact of ASCT, transplant refusal and high-risk MM. Bone Marrow Transplant 2024; 59:660-669. [PMID: 38383715 PMCID: PMC11073964 DOI: 10.1038/s41409-024-02227-0] [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: 07/22/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
In Hong Kong, newly diagnosed multiple myeloma (NDMM) receives bortezomib-based triplet induction. Upfront autologous stem cell transplant (ASCT) is offered to transplant eligible (TE) patients (NDMM ≤ 65 years of age), unless medically unfit (TE-unfit) or refused (TE-refused). Data was retrieved for 448 patients to assess outcomes. For the entire cohort, multivariate analysis showed that male gender (p = 0.006), international staging system (ISS) 3 (p = 0.003), high lactate dehydrogenase (LDH) (p = 7.6 × 10-7) were adverse predictors for overall survival (OS), while complete response/ near complete response (CR/nCR) post-induction (p = 2.7 × 10-5) and ASCT (p = 4.8 × 10-4) were favorable factors for OS. In TE group, upfront ASCT was conducted in 252 (76.1%). Failure to undergo ASCT in TE patients rendered an inferior OS (TE-unfit p = 1.06 × 10-8, TE-refused p = 0.002) and event free survival (EFS) (TE-unfit p = 0.00013, TE-refused p = 0.002). Among TE patients with ASCT, multivariate analysis showed that age ≥ 60 (p = 8.9 × 10-4), ISS 3 (p = 0.019) and high LDH (p = 2.6 × 10-4) were adverse factors for OS. In those with high-risk features (HR cytogenetics, ISS 3, R-ISS 3), ASCT appeared to mitigate their adverse impact. Our data reaffirmed the importance of ASCT. The poor survival inherent with refusal of ASCT should be recognized by clinicians. Finally, improved outcome with ASCT in those with high-risk features warrant further studies.
Collapse
Affiliation(s)
- Hoi Ki Karen Tang
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Chi Yeung Fung
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yu Yan Hwang
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Harold Lee
- Department of Medicine, Princess Margaret Hospital, Kwai Chung, Hong Kong
| | - Grace Lau
- Department of Medicine, Princess Margaret Hospital, Kwai Chung, Hong Kong
| | - Sze Fai Yip
- Department of Medicine, Tuen Mun Hospital, Tuen Mun, Hong Kong
| | - Bonnie Kho
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Chai Wan, Hong Kong
| | - Chi Kuen Lau
- Department of Medicine, Tseung Kwan O Hospital, Tseung Kwan O, Hong Kong
| | - Kwan Hung Leung
- Department of Medicine, United Christian Hospital, Kwun Tong, Hong Kong
| | - Elaine Au
- Department of Pathology, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Eric Tse
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Joycelyn Sim
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yok Lam Kwong
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, University of Hong Kong, Pok Fu Lam, Hong Kong.
- Department of Medicine, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong.
| |
Collapse
|
129
|
Dimopoulos M, Sonneveld P, Manier S, Lam A, Roccia T, Schecter JM, Cost P, Pacaud L, Poirier A, Tremblay G, Lan T, Valluri S, Kumar S. Progression-free survival as a surrogate endpoint for overall survival in patients with relapsed or refractory multiple myeloma. BMC Cancer 2024; 24:541. [PMID: 38684948 PMCID: PMC11057089 DOI: 10.1186/s12885-024-12263-0] [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: 12/15/2023] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
OBJECTIVES The goal of the research was to assess the quantitative relationship between median progression-free survival (PFS) and median overall survival (OS) specifically among patients with relapsed/refractory multiple myeloma (RRMM) based on published randomized controlled trials (RCTs). METHODS Two bibliographic databases (PubMed and Embase, 1970-2017) were systematically searched for RCTs in RRMM that reported OS and PFS, followed by an updated search of studies published between 2010 and 2022 in 3 databases (Embase, MEDLINE, and EBM Reviews, 2010-2022). The association between median PFS and median OS was assessed using the nonparametric Spearman rank and parametric Pearson correlation coefficients. Subsequently, the quantitative relationship between PFS and OS was assessed using weighted least-squares regression adjusted for covariates including age, sex, and publication year. Study arms were weighted by the number of patients in each arm. RESULTS A total of 31 RCTs (56 treatment arms, 10,450 patients with RRMM) were included in the analysis. The average median PFS and median OS were 7.1 months (SD 5.5) and 28.1 months (SD 11.8), respectively. The Spearman and Pearson correlation coefficients between median PFS and median OS were 0.80 (P < 0.0001) and 0.79 (P < 0.0001), respectively. In individual treatment arms of RRMM trials, each 1-month increase in median PFS was associated with a 1.72-month (95% CI 1.26-2.17) increase in median OS. CONCLUSION Analysis of the relationship between PFS and OS incorporating more recent studies in RRMM further substantiates the use of PFS to predict OS in RRMM.
Collapse
Affiliation(s)
- Meletios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | | | - Abbey Poirier
- Cytel Inc. Health Economics & Outcomes Research, Waltham, MA, USA
| | - Gabriel Tremblay
- Cytel Inc. Health Economics & Outcomes Research, Waltham, MA, USA
| | - Tommy Lan
- Cytel Inc. Health Economics & Outcomes Research, Waltham, MA, USA
| | | | - Shaji Kumar
- Department of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
130
|
Takahashi S, Minnie SA, Ensbey KS, Schmidt CR, Sekiguchi T, Legg SRW, Zhang P, Koyama M, Olver SD, Collinge AD, Keshmiri S, Comstock ML, Varelias A, Green DJ, Hill GR. Regulatory T cells suppress myeloma-specific immunity during autologous stem cell mobilization and transplantation. Blood 2024; 143:1656-1669. [PMID: 38295333 PMCID: PMC11103090 DOI: 10.1182/blood.2023022000] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/02/2024] Open
Abstract
ABSTRACT Autologous stem cell transplantation (ASCT) is the standard of care consolidation therapy for eligible patients with myeloma but most patients eventually progress, an event associated with features of immune escape. Novel approaches to enhance antimyeloma immunity after ASCT represent a major unmet need. Here, we demonstrate that patient-mobilized stem cell grafts contain high numbers of effector CD8 T cells and immunosuppressive regulatory T cells (Tregs). We showed that bone marrow (BM)-residing T cells are efficiently mobilized during stem cell mobilization (SCM) and hypothesized that mobilized and highly suppressive BM-derived Tregs might limit antimyeloma immunity during SCM. Thus, we performed ASCT in a preclinical myeloma model with or without stringent Treg depletion during SCM. Treg depletion generated SCM grafts containing polyfunctional CD8 T effector memory cells, which dramatically enhanced myeloma control after ASCT. Thus, we explored clinically tractable translational approaches to mimic this scenario. Antibody-based approaches resulted in only partial Treg depletion and were inadequate to recapitulate this effect. In contrast, a synthetic interleukin-2 (IL-2)/IL-15 mimetic that stimulates the IL-2 receptor on CD8 T cells without binding to the high-affinity IL-2Ra used by Tregs efficiently expanded polyfunctional CD8 T cells in mobilized grafts and protected recipients from myeloma progression after ASCT. We confirmed that Treg depletion during stem cell mobilization can mitigate constraints on tumor immunity and result in profound myeloma control after ASCT. Direct and selective cytokine signaling of CD8 T cells can recapitulate this effect and represent a clinically testable strategy to improve responses after ASCT.
Collapse
Affiliation(s)
- Shuichiro Takahashi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Simone A. Minnie
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Kathleen S. Ensbey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Christine R. Schmidt
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Tomoko Sekiguchi
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Samuel R. W. Legg
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Ping Zhang
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Motoko Koyama
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Stuart D. Olver
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | | | - Sara Keshmiri
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Melissa L. Comstock
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, St Lucia, QLD, Australia
| | - Damian J. Green
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Geoffrey R. Hill
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
131
|
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
|
132
|
Maura F, Rajanna AR, Ziccheddu B, Poos AM, Derkach A, Maclachlan K, Durante M, Diamond B, Papadimitriou M, Davies F, Boyle EM, Walker B, Hultcrantz M, Silva A, Hampton O, Teer JK, Siegel EM, Bolli N, Jackson GH, Kaiser M, Pawlyn C, Cook G, Kazandjian D, Stein C, Chesi M, Bergsagel L, Mai EK, Goldschmidt H, Weisel KC, Fenk R, Raab MS, Van Rhee F, Usmani S, Shain KH, Weinhold N, Morgan G, Landgren O. Genomic Classification and Individualized Prognosis in Multiple Myeloma. J Clin Oncol 2024; 42:1229-1240. [PMID: 38194610 PMCID: PMC11095887 DOI: 10.1200/jco.23.01277] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/08/2023] [Accepted: 10/23/2023] [Indexed: 01/11/2024] Open
Abstract
PURPOSE Outcomes for patients with newly diagnosed multiple myeloma (NDMM) are heterogenous, with overall survival (OS) ranging from months to over 10 years. METHODS To decipher and predict the molecular and clinical heterogeneity of NDMM, we assembled a series of 1,933 patients with available clinical, genomic, and therapeutic data. RESULTS Leveraging a comprehensive catalog of genomic drivers, we identified 12 groups, expanding on previous gene expression-based molecular classifications. To build a model predicting individualized risk in NDMM (IRMMa), we integrated clinical, genomic, and treatment variables. To correct for time-dependent variables, including high-dose melphalan followed by autologous stem-cell transplantation (HDM-ASCT), and maintenance therapy, a multi-state model was designed. The IRMMa model accuracy was significantly higher than all comparator prognostic models, with a c-index for OS of 0.726, compared with International Staging System (ISS; 0.61), revised-ISS (0.572), and R2-ISS (0.625). Integral to model accuracy was 20 genomic features, including 1q21 gain/amp, del 1p, TP53 loss, NSD2 translocations, APOBEC mutational signatures, and copy-number signatures (reflecting the complex structural variant chromothripsis). IRMMa accuracy and superiority compared with other prognostic models were validated on 256 patients enrolled in the GMMG-HD6 (ClinicalTrials.gov identifier: NCT02495922) clinical trial. Individualized patient risks were significantly affected across the 12 genomic groups by different treatment strategies (ie, treatment variance), which was used to identify patients for whom HDM-ASCT is particularly effective versus patients for whom the impact is limited. CONCLUSION Integrating clinical, demographic, genomic, and therapeutic data, to our knowledge, we have developed the first individualized risk-prediction model enabling personally tailored therapeutic decisions for patients with NDMM.
Collapse
Affiliation(s)
- Francesco Maura
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Arjun Raj Rajanna
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Bachisio Ziccheddu
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Alexandra M. Poos
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andriy Derkach
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Durante
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Benjamin Diamond
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Marios Papadimitriou
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Faith Davies
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Eileen M. Boyle
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Brian Walker
- Division of Hematology Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ariosto Silva
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | | | - Jamie K. Teer
- Department of Biostatistics & Bioinformatics, Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Erin M. Siegel
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL
| | - Niccolò Bolli
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Graham H. Jackson
- Freeman Hospital, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - Martin Kaiser
- The Institute of Cancer Research, London, United Kingdom
| | - Charlotte Pawlyn
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Gordon Cook
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Dickran Kazandjian
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Caleb Stein
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Marta Chesi
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Leif Bergsagel
- Division of Hematology/Oncology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Elias K. Mai
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Katja C. Weisel
- Department of Oncology, Hematology and Blood and Marrow Transplant, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Fenk
- Department of Hematology, Oncology and Clinical Immunology, University-Hospital Duesseldorf, Duesseldorf, Germany
| | - Marc S. Raab
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fritz Van Rhee
- Myeloma Institute for Research & Therapy, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Saad Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kenneth H. Shain
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Niels Weinhold
- Heidelberg Myeloma Center, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit (CCU) Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gareth Morgan
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
133
|
Li CC, Tsai XCH, Huang WH, Wang TF. Recent advancements in hematopoietic stem cell transplantation in Taiwan. Tzu Chi Med J 2024; 36:127-135. [PMID: 38645784 PMCID: PMC11025591 DOI: 10.4103/tcmj.tcmj_276_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 04/23/2024] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) can cure malignant and nonmalignant hematological disorders. From 1983 to 2022, Taiwan performed more than 10,000 HSCT transplants. The Taiwan Blood and Marrow Transplantation Registry collects clinical information to gather everyone's experience and promote the advances of HSCT in Taiwan to gather everyone's experience and promote advances of HSCT in Taiwan. Compared with matched sibling donors, transplants from matched unrelated donors exhibited a trend of superior survival. In Taiwan, transplant donors showed remarkable growth from unrelated (24.8%) and haploidentical (10.5%) donors. The number of older patients (17.4%; aged ≥61 years) who underwent transplantation has increased markedly. This review summarizes several significant developments in HSCT treatment in Taiwan. First, the use of Anti-thymocyte globulin (ATG) and intravenous busulfan regimens were important risk factors for predicting hepatic sinusoidal obstruction syndrome. Second, a new, machine learning-based risk prediction scoring system for posttransplantation lymphoproliferative disorder has identified five risk factors: aplastic anemia, partially mismatched related donors, fludarabine use, ATG use, and acute skin graft-versus-host disease. Third, although the incidence of idiopathic pneumonia syndrome was low (1.1%), its mortality rate was high (58.1%). Fourth, difficult-to-treat mantle cell and T-cell lymphomas treated with autologous HSCT during earlier remission had higher survival rates. Fifth, treatment of incurable multiple myeloma with autologous HSCT showed a median progression-free survival and overall survival of 46.5 and 70.4 months, respectively. Sixth, different haploidentical transplantation strategies were compared. Seventh, caution should be taken in administering allogeneic HSCT treatment in older patients with myeloid leukemia with a Charlson Comorbidity Index ≥3 because of a higher risk of nonrelapse mortality.
Collapse
Affiliation(s)
- Chi-Cheng Li
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of Stem Cell and Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Xavier Cheng-Hong Tsai
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
- Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Wei-Han Huang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of Stem Cell and Precision Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Clinical Pathology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tso-Fu Wang
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
134
|
Ravi G, Bal S, Joiner L, Giri S, Sentell M, Hill T, Godby KN, Costa LJ. Subsequent therapy and outcomes in patients with newly diagnosed multiple myeloma experiencing disease progression after quadruplet combinations. Br J Haematol 2024; 204:1300-1306. [PMID: 38291707 DOI: 10.1111/bjh.19303] [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: 11/22/2023] [Revised: 12/24/2023] [Accepted: 01/07/2024] [Indexed: 02/01/2024]
Abstract
The combination of anti-CD38 monoclonal antibodies to a proteasome inhibitor, an immunomodulatory agent and dexamethasone (quadruplet-QUAD) in sequence with autologous stem cell transplantation (ASCT) leads to deep and durable responses in newly diagnosed multiple myeloma (NDMM). Disease progression in the first year post-QUADs is uncommon. We analysed 274 consecutive NDMM patients treated with QUADs + ASCT. After a median follow-up of 21.3 months, 20 patients had disease progression <18 months and 21 had progression ≥18 months after the onset of a QUAD regimen. All patients received subsequent anti-MM therapy, and 38 were evaluated for response. Nine (22.0%) received T-cell redirecting therapy as the next treatment, and 21 (51.2%) at some point in the treatment course. Response to next therapy was 26.3% for patients with progression <18 months and 52.6% for those with progression ≥18 months after the onset of a QUAD regimen. Median PFS on the next therapy was 2.5 months (95% CI 1.5-3.4) for those with progression <18 months and 7.0 months (95% CI 3.6-10.5) for those with progression ≥18 months. Efforts should focus on the early deployment of therapies with new mechanism of action for patients experiencing treatment failure after QUADs.
Collapse
Affiliation(s)
- Gayathri Ravi
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Susan Bal
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Laura Joiner
- Department of Pharmacy, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Smit Giri
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melissa Sentell
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Tiffany Hill
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kelly N Godby
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Luciano J Costa
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
135
|
Guare EG, Hale CM, Sivik J, Lehman E, Inoue Y, Rakszawski K, Songdej N, Nickolich M, Zheng H, Naik S, Claxton D, Rybka W, Hohl R, Mineishi S, Minagawa K, Paules CI. The addition of doxycycline to fluoroquinolones for bacterial prophylaxis in autologous stem cell transplantation for multiple myeloma. Transpl Infect Dis 2024; 26:e14241. [PMID: 38269469 DOI: 10.1111/tid.14241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Bacterial prophylaxis with a fluoroquinolone (FQ) during autologous stem cell transplant (ASCT) is common, although not standardized among transplant centers. The addition of doxycycline (doxy) to FQ prophylaxis was previously linked to reduced neutropenic fever and bacteremia in multiple myeloma (MM) patients undergoing ASCT although several confounders were present. We compared the incidence of neutropenic fever and bacteremia between MM patients variably receiving prophylaxis with FQ alone and FQ-doxy during ASCT. METHODS Systematic retrospective chart review of MM patients who underwent ASCT between January 2016 and December 2021. The primary objective was to determine the effect of bacterial prophylaxis on neutropenic fever and bacteremia within 30 days of ASCT. Multivariable logistic regression for neutropenic fever and univariate logistic regression for bacteremia accounted for differences in subject characteristics between groups. RESULTS Among 341 subjects, 121 received FQ and 220 received FQ-doxy for prophylaxis. Neutropenic fever developed in 67 (55.4%) and 87 (39.5%) subjects in the FQ and FQ-doxy groups, respectively (p = .005). Bacteremia was infrequent, with 5 (4.1%) and 5 (2.3%) cases developing in the FQ and FQ-doxy groups, respectively (p = .337). Among Gram-negative bacteremia events, 7/7 Escherichia coli strains were FQ-resistant, and 5/7 were ceftriaxone-resistant. CONCLUSION The FQ-doxy prophylaxis group had fewer cases of neutropenic fever than the FQ group, however, there was no significant difference in bacteremia. High rates of antibiotic resistance were observed. An updated randomized controlled trial investigating appropriate prophylaxis for ASCT in the context of current oncology standards and changing antimicrobial resistance rates is warranted.
Collapse
Affiliation(s)
- Emma G Guare
- Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Cory M Hale
- Department of Pharmacy, Penn State Health Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Jeffrey Sivik
- Department of Pharmacy, Penn State Health Milton S. Hershey Medical Center and Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Erik Lehman
- Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Yoshika Inoue
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Kevin Rakszawski
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Natthapol Songdej
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Myles Nickolich
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Hong Zheng
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Seema Naik
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - David Claxton
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Witold Rybka
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Raymond Hohl
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Shin Mineishi
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Kentaro Minagawa
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, Pennsylvania, USA
| | - Catharine I Paules
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
136
|
Abstract
No therapy in multiple myeloma has been as extensively investigated as stem cell transplantation following high-dose chemotherapy. A search of the national library of medicine in February 2023 revealed over 27,000 publications covering stem cell transplantation. No other treatment for multiple myeloma has been so vigorously investigated. However, given the rapid advances seen in the treatment of multiple myeloma, it is legitimate to ask whether the technique first introduced in 1983 by Thomas McIlwain still has relevance. In 1984,Barlogie introduced infusional vincristine, doxorubicin, and dexamethasone and in 1986 published a first series on high-dose therapy with autologous marrow-derived stem cells. At this point, the only available therapies were melphalan, prednisone, other intensive steroids such as methylprednisolone, and interferon. Cyclophosphamide was used both orally and parenterally. VBMCP was introduced as a combination therapy at Memorial Hospital subsequently shown not to be superior to melphalan and prednisone.
Collapse
Affiliation(s)
- Morie A Gertz
- Mayo Clinic, 200 SW First street, Rochester, MN 55905, USA.
| |
Collapse
|
137
|
Neuendorff NR, Khan A, Ullrich F, Yates S, Devarakonda S, Lin RJ, von Tresckow B, Cordoba R, Artz A, Rosko AE. Cellular therapies in older adults with hematological malignancies: A case-based, state-of-the-art review. J Geriatr Oncol 2024; 15:101734. [PMID: 38430810 DOI: 10.1016/j.jgo.2024.101734] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/05/2023] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Cellular therapies, including autologous stem cell transplant (ASCT), allogeneic hematopoietic cell transplantation (alloHCT), and chimeric antigen receptor- (CAR-) T cell therapies are essential treatment modalities for many hematological malignancies. Although their use in older adults has substantially increased within the past decades, cellular therapies represent intensive treatment approaches that exclude a large percentage of older adults due to comorbidities and frailty. Under- and overtreatment in older adults with hematologic malignancy is a challenge and many treatment decisions are influenced by chronologic age. The advent of efficient and well-tolerated newer treatment approaches for multiple myeloma has challenged the role of ASCT. In the modern era, there are no randomized clinical trials of transplant versus non-transplant strategies for patients ≥65 years. Nonetheless, ASCT is feasible for selected older patients and does not result in long-term compromise in quality of life. AlloHCT is the only curative approach for acute myeloid leukemia of intermediate and unfavourable risk but carries a significant risk for non-relapse mortality depending on comorbidities, general fitness, and transplant-specific characteristics, such as intensity of conditioning and donor choice. However, alloHCT is feasible in appropriately-selected older adults. Early referral for evaluation is strongly encouraged as this is the most obvious barrier. CAR-T cell therapies have shown unprecedented clinical efficacy and durability in relapsed and refractory diffuse large B cell lymphoma. Its use is well tolerated in older adults, although evidence comes from limited case numbers. Whether patients who are deemed unfit for ASCT qualify for CAR-T cell therapy remains elusive, but the tolerability and efficacy of CAR-T cell therapy appears promising, especially for older patients. The evidence from randomized trials is strong in favor of using a comprehensive geriatric assessment (CGA) to reduce treatment-related toxicities and guide treatment intensity in the care for solid tumors; its use for evaluation of cellular therapies is less evidence-based. However, CGA can provide useful information on patients' fitness, resilient mechanisms, and reveal potential optimization strategies for compensating for vulnerabilities. In this narrative review, we will discuss key questions on cellular therapies in older adults based on illustrative patient cases.
Collapse
Affiliation(s)
- Nina Rosa Neuendorff
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, D-45147 Essen, Germany.
| | - Abdullah Khan
- Department of Hematology, The Ohio State University, James Comprehensive Cancer Center, Columbus, OH, United States of America
| | - Fabian Ullrich
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, D-45147 Essen, Germany
| | - Samuel Yates
- Department of Internal Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, United States of America
| | - Srinivas Devarakonda
- Department of Hematology, The Ohio State University, James Comprehensive Cancer Center, Columbus, OH, United States of America
| | - Richard J Lin
- Adult Bone Marrow Transplantation (BMT) Service, Cellular Therapy Service, Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, D-45147 Essen, Germany
| | - Raul Cordoba
- Lymphoma Unit, Department of Hematology, Health Research Institute IIS-FJD, Fundacion Jimenez Diaz University Hospital, Madrid, Spain
| | - Andrew Artz
- Division of Leukemia, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, Duarte, CA, USA
| | - Ashley E Rosko
- Department of Hematology, The Ohio State University, James Comprehensive Cancer Center, Columbus, OH, United States of America
| |
Collapse
|
138
|
Salafian K, Mazimba C, Volodin L, Varadarajan I, Pilehvari A, You W, Knio ZO, Ballen K. The impact of social vulnerability index on survival following autologous stem cell transplant for multiple myeloma. Bone Marrow Transplant 2024; 59:459-465. [PMID: 38238453 PMCID: PMC10994832 DOI: 10.1038/s41409-024-02200-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/29/2023] [Accepted: 01/05/2024] [Indexed: 04/06/2024]
Abstract
Autologous hematopoietic stem cell transplantation (ASCT) is the standard of care for eligible patients with multiple myeloma (MM) to prolong progression-free survival (PFS). While several factors affect survival following ASCT, the impact of social determinants of health such as the CDC Social Vulnerability Index (SVI) is not well documented. This single-center retrospective analysis evaluated the impact of SVI on PFS following ASCT in MM patients. 225 patients with MM who underwent ASCT participated, with 51% transplanted in the last 5 years. At 5 years post-transplant, 55 (50%) achieved PFS and 66 (60%) remained alive. Higher SVI values were significantly associated with lower odds of PFS (OR = 0.521, p < 0.01, 95% CI [0.41, 0.66]) and OS (OR = 0.592, p < 0.01, 95% CI [0.46, 0.76]) post-transplant. Greater vulnerability scores in the socioeconomic status (OR = 0.890; 95% CI: [0.82, 0.96]), household characteristics (OR = 0.912; 95% CI: [0.87, 0.95]), and racial and ethnic minority status (OR = 0.854; 95% CI: [0.81, 0.90]) themes significantly worsened the odds of PFS. These results suggest high SVI areas may need more resources to achieve optimal PFS and OS. Future studies will focus on addressing factors within the socioeconomic status, household characteristics, and racial and ethnic minority subthemes, as these have a more pronounced effect on PFS.
Collapse
Affiliation(s)
- Kiarash Salafian
- Department of Medicine, University of Virginia Health, Charlottesville, VA, USA
| | - Christine Mazimba
- Division of Hematology/Oncology, University of Virginia Health, Charlottesville, VA, USA
| | - Leonid Volodin
- Division of Hematology/Oncology, University of Virginia Health, Charlottesville, VA, USA
| | - Indumathy Varadarajan
- Division of Hematology/Oncology, University of Virginia Health, Charlottesville, VA, USA
| | - Asal Pilehvari
- Department of Public Health Sciences, University of Virginia, and University of Virginia Comprehensive Cancer Center, Charlottesville, VA, USA
| | - Wen You
- Department of Public Health Sciences, University of Virginia, and University of Virginia Comprehensive Cancer Center, Charlottesville, VA, USA
| | - Ziyad O Knio
- Department of Anesthesiology, University of Virginia Health, Charlottesville, VA, USA
| | - Karen Ballen
- Division of Hematology/Oncology, University of Virginia Health, Charlottesville, VA, USA.
| |
Collapse
|
139
|
Brazauskas R, Flynn K, Krishnan A, Landau H, Giralt S, Pasquini MC, Stadtmauer EA, D’Souza A. Symptom clusters and their impact on quality of life in multiple myeloma survivors: Secondary analysis of BMT CTN 0702 trial. Br J Haematol 2024; 204:1429-1438. [PMID: 38348544 PMCID: PMC11006567 DOI: 10.1111/bjh.19326] [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: 10/19/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 04/11/2024]
Abstract
Autologous haematopoietic cell transplantation (autoHCT) and continuous post-transplant maintenance therapy are the standard of care in transplant-eligible multiple myeloma (MM) patients. We sought to describe symptom burden and identify symptom clusters occurring in MM patients after autoHCT using data from the BMT CTN 0702 randomized controlled trial comparing the outcomes of three treatment interventions after an autoHCT in 758 MM patients. We analysed individual transplant-related symptoms assessed via the FACT-BMT questionnaire at enrolment and annually for 4-year post-autoHCT. We also described the effect the individual symptoms and symptom clusters have on quality of life (QoL). We identified three stable symptom clusters: malaise symptom cluster (lack of energy, feeling ill, having pain, experiencing nausea, loss of appetite), physical symptom cluster (having skin problems, tremors, worsening eyesight, change in taste, shortness of breath, frequent colds) and emotional symptom cluster (feeling sad, being nervous, experiencing sleep problems). Malaise and emotional symptom clusters have a greater impact on QoL than the physical symptoms cluster. Identifying these symptoms warrant additional support in terms of psychosocial support, in addition to treatment of the physical symptoms themselves.
Collapse
Affiliation(s)
- Ruta Brazauskas
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kathryn Flynn
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope Cancer Center, Duarte, CA, USA
| | - Heather Landau
- Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio Giralt
- Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcelo C Pasquini
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Edward A Stadtmauer
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anita D’Souza
- Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
140
|
Devasia AJ, Lancman GS, Stewart AK. Management of Newly Diagnosed Multiple Myeloma Today, and in the Future. Hematol Oncol Clin North Am 2024; 38:441-459. [PMID: 38171937 DOI: 10.1016/j.hoc.2023.12.007] [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: 01/05/2024]
Abstract
Treatment options have expanded rapidly and widely in the past two decades for patients with multiple myeloma. Triplet novel agent-based induction regimens have been accepted as the standard practice wordwide over the last decade both for transplant-eligible and non-eligible patients. The addition of anti-CD38 monoclonal antibodies as part of quadruplet regimens has led to even deeper and longer-lasting responses. The impressive results shown by the quadruplets havebeen practice-changing where accessible in recent years. Chimeric antigen receptor T cell therapy and bispecific antibodies are being tested in the upfront setting and have the potential to once again shift the paradigm of treatment of newly diagnosed MM.
Collapse
Affiliation(s)
- Anup Joseph Devasia
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, 700 University Avenue, Toronto, Ontario, M5G 1X6 Canada
| | - Guido Sebastian Lancman
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, 700 University Avenue, Toronto, Ontario, M5G 1X6 Canada
| | - Alexander Keith Stewart
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, 700 University Avenue, Toronto, Ontario, M5G 1X6 Canada.
| |
Collapse
|
141
|
Beksac M, Eikema DJ, Koster L, Hulin C, Poiré X, Hamladji RM, Gromek T, Bazarbachi A, Ozkurt ZN, Pabst T, Ben Othman T, Finke J, Pirogova O, Wu D, Hayat A, Hilgendorf I, Tholouli E, de Wreede LC, Schönland S, Garderet L, Drozd-Sokolowska J, Raj K, Hayden PJ, Yakoub-Agha I, McLornan DP. In the era of Bortezomib-based Induction, intensification of Melphalan-based conditioning with Bortezomib does not improve Survival Outcomes in newly diagnosed Multiple Myeloma: a study from the Chronic Malignancies Working Party of the EBMT. Bone Marrow Transplant 2024; 59:526-533. [PMID: 38297040 PMCID: PMC10994834 DOI: 10.1038/s41409-023-02160-8] [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: 09/04/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 02/02/2024]
Abstract
Bortezomib (Vel)- Melphalan 200 mg/m2 (Mel200) (Vel-Mel) has been utilised to intensify conditioning in autologous hematopoietic stem cell transplantation (AHCT) for multiple myeloma (MM). This EBMT registry-based study compared Vel-Mel with Mel200 during upfront AHCT. Between 2010 and 2017, MM patients who received Vel-Mel (n = 292) conditioning were compared with 4,096 Mel200 patients in the same 58 centres. Pre-AHCT, compared to Mel200 patients, Vel-Mel patients had similar International Staging System (ISS) scores and cytogenetic risk profiles; a similar proportion had received bortezomib-based induction (85% and 87.3%, respectively) though they were younger with a better performance status. Vel-Mel patients were more likely to achieve CR post-induction (40.6% vs 20.3%, p < 0.001) and by day 100 of AHCT (CR/VGPR: 70.2 % vs. 57.2%, p < 0.001). There was no difference in 3-year PFS (49% vs 46%, p = 0.06) or early post-AHCT mortality. In multivariable analysis, Vel-Mel associated with inferior PFS (HR: 1.69 (1.27-2.25, p < 0.001) and OS (HR:1.46 (1.14-1.86,p = 0.002), similar to negative effects on PFS of advanced ISS (HR:1.56 (1.33-1.83, p < 0.001), high-risk cytogenetics (HR:1.43(1.18-1.74, p < 0.001) and poor post-induction response(<=PR)(HR: 1.43(1.25-1.62, p < 0.001) Overall, despite superior pre- and post-AHCT responses, there was no improvement in PFS or OS following Vel-Mel. This data supports the findings of the smaller prospective IFM study.
Collapse
Affiliation(s)
- Meral Beksac
- Istinye University Ankara Liv Hospital Hematology and Stem Cell Transplantation Unit, Ankara, 06880, Turkey.
| | | | | | | | - Xavier Poiré
- Cliniques Universitaires St. Luc, Brussels, Belgium
| | | | | | - Ali Bazarbachi
- Bone Marrow Transplantation Program, American University of Beirut Medical Center, Beiruit, Lebanon
| | | | | | | | | | - Olga Pirogova
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russian Federation
| | - Depei Wu
- First Affiliated Hospital of Soochow University, Suzhou, China
| | - Amjad Hayat
- The Blood and Tissue Establishment, Galway University Hospital, Galway, Ireland
| | | | | | - Liesbeth C de Wreede
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Stefan Schönland
- Medizinische Klinik u. Poliklinik V, University of Heidelberg, Heidelberg, Germany
| | | | | | - Kavita Raj
- Department of Stem Cell Transplantation, University College London Hospitals, London, UK
| | - Patrick J Hayden
- Department of Haematology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | | | - Donal P McLornan
- Department of Stem Cell Transplantation, University College London Hospitals, London, UK
| |
Collapse
|
142
|
Hwang A, Hayden P, Pawlyn C, McLornan D, Garderet L. The role of maintenance therapy following autologous stem cell transplantation in newly diagnosed multiple myeloma: Considerations on behalf of the Chronic Malignancies Working Party of the EBMT. Br J Haematol 2024; 204:1159-1175. [PMID: 38390784 DOI: 10.1111/bjh.19353] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/22/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
Recent treatment advancements in multiple myeloma have led to significant improvements in patient outcomes. Maintenance therapy following autologous haematopoietic stem cell transplantation (AHCT) is now standard of care and has been demonstrated to prolong and deepen treatment responses. Currently, lenalidomide remains the single agent that has been approved for maintenance post-AHCT in Europe and the USA which, if tolerated, is continued until disease progression. The treatment landscape is rapidly expanding however, and the optimal personalised maintenance approach for a patient is becoming more complex. Treatment outcomes for patients with high-risk disease remain poor and choice of maintenance in this population also remains unclear. This review article evaluates up-to-date literature regarding established maintenance approaches. It further analyses ongoing studies exploring maintenance regimens using combination and novel agents, approaches to maintenance in patients with cytogenetic high-risk disease and minimal residual disease response-adapted strategies that reflect the current evolving treatment paradigm.
Collapse
Affiliation(s)
- Angela Hwang
- Department of Haematology, University College London Hospital NHS Foundation Trust, London, UK
| | - Patrick Hayden
- Department of Haematology, St James's Hospital, Dublin, Ireland
| | | | - Donal McLornan
- Department of Haematology, University College London Hospital NHS Foundation Trust, London, UK
| | - Laurent Garderet
- Département d'Hématologie, Hôpital Pitié Salpêtrière, AP-HP, Paris, France
| |
Collapse
|
143
|
Hughes CFM, Shah GL, Paul BA. Autologous hematopoietic stem cell transplantation for multiple myeloma in the age of CAR T cell therapy. Front Oncol 2024; 14:1373548. [PMID: 38601770 PMCID: PMC11004402 DOI: 10.3389/fonc.2024.1373548] [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: 01/19/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the management of relapsed and refractory myeloma, with excellent outcomes and a tolerable safety profile. High dose chemotherapy with autologous hematopoietic stem cell transplantation (AHCT) is established as a mainstream of newly diagnosed multiple myeloma (NDMM) management in patients who are young and fit enough to tolerate such intensity. This standard was developed based on randomized trials comparing AHCT to chemotherapy in the era prior to novel agents. More recently, larger studies have primarily shown a progression free survival (PFS) benefit of upfront AHCT, rather than overall survival (OS) benefit. There is debate about the significance of this lack of OS, acknowledging the potential confounders of the chronic nature of the disease, study design and competing harms and benefits of exposure to AHCT. Indeed upfront AHCT may not be as uniquely beneficial as we once thought, and is not without risk. New quadruple-agent regimens are highly active and effective in achieving a deep response as quantified by measurable residual disease (MRD). The high dose chemotherapy administered with AHCT imposes a burden of short and long-term adverse effects, which may alter the disease course and patient's ability to tolerate future therapies. Some high-risk subgroups may have a more valuable benefit from AHCT, though still ultimately suffer poor outcomes. When compared to the outcomes of CAR T cell therapy, the question of whether AHCT can or indeed should be deferred has become an important topic in the field. Deferring AHCT may be a personalized decision in patients who achieve MRD negativity, which is now well established as a key prognostic factor for PFS and OS. Reserving or re-administering AHCT at relapse is feasible in many cases and holds the promise of resetting the T cell compartment and opening up options for immune reengagement. It is likely that personalized MRD-guided decision making will shape how we sequence in the future, though more studies are required to delineate when this is safe and appropriate.
Collapse
Affiliation(s)
- Charlotte F. M. Hughes
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Gunjan L. Shah
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Barry A. Paul
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health/Wake Forest Baptist, Charlotte, NC, United States
| |
Collapse
|
144
|
Rafae A, van Rhee F, Al Hadidi S. Perspectives on the Treatment of Multiple Myeloma. Oncologist 2024; 29:200-212. [PMID: 37995307 PMCID: PMC10911930 DOI: 10.1093/oncolo/oyad306] [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: 05/24/2023] [Accepted: 10/29/2023] [Indexed: 11/25/2023] Open
Abstract
The treatment of multiple myeloma has evolved significantly over the past few decades with the development of novel therapeutics. The introduction of proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, and high-dose chemotherapy followed by hematopoietic stem cell transplantation has led to improved response rates and survival outcomes. The use of bispecific antibodies and chimeric antigen receptor T-cell therapy is currently under study, and early results are showing promise. Although outcomes for patients with MM have improved with the development of new treatments, there remains a subset of patients with high-risk disease who have a particularly poor prognosis. Therefore, it is critical that future clinical trials focus on developing new therapies specifically for high-risk multiple myeloma. Here we review the literature and provide guidance on treating patients with multiple myeloma for practicing oncologists.
Collapse
Affiliation(s)
- Abdul Rafae
- Department of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Frits van Rhee
- Myeloma Institute, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Samer Al Hadidi
- Myeloma Institute, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
145
|
Hammons L, Haider S, Portuguese AJ, Banerjee R, Szabo A, Pasquini M, Chhabra S, Radhakrishnan S, Mohan M, Narra R, Dong J, Janz S, Shah NN, Hamadani M, D'Souza A, Hari P, Dhakal B. Chimeric antigen receptor and bispecific T-cell engager therapies in multiple myeloma patients with prior allogeneic transplantation. Br J Haematol 2024; 204:887-891. [PMID: 38054558 DOI: 10.1111/bjh.19244] [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: 08/31/2023] [Revised: 11/08/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy and bispecific T-cell engagers (BsAb) have emerged as promising immunotherapeutic modalities in patients with relapsed and/or refractory multiple myeloma (RRMM). However, there is limited data on the safety and efficacy of CAR-T and BsAb therapies in MM patients with a prior history of allogeneic transplantation (allo-HCT). Thirty-three MM patients with prior allo-HCT received CAR-T (n = 24) or BsAb (n = 9) therapy. CAR-T therapy demonstrated an ORR of 92% (67% ≥ CR), and 73% were MRD negative. BsAb therapy resulted in an ORR of 44% (44% ≥ CR) and 44% MRD negative. Safety analysis showed grade ≥3 AEs in 92% of CAR-T and 56% of BsAb patients. Cytokine release syndrome (CRS) occurred in 83% of CAR-T and 78% of BsAb recipients, while immune effector cell-associated neurotoxicity syndrome (ICANS) was observed in three CAR-T patients. Infections of grade ≥3 were reported in 50% of CAR-T and 44% of BsAb recipients. No exacerbation of graft-versus-host disease occurred except in one BsAb recipient. CAR-T and BsAb therapies appear to be feasible, safe and provide deep and durable responses in MM patients with prior allo-HCT.
Collapse
Affiliation(s)
- Lindsay Hammons
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Shabi Haider
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andrew J Portuguese
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rahul Banerjee
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Aniko Szabo
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Marcelo Pasquini
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Saurabh Chhabra
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic Arizona, Phoenix, Arizona, USA
| | - Sabarinath Radhakrishnan
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Meera Mohan
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ravi Narra
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jing Dong
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Siegfried Janz
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Nirav N Shah
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Mehdi Hamadani
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Anita D'Souza
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Parameswaran Hari
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Binod Dhakal
- BMT and Cellular Therapy Program, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
146
|
Shah G, Giralt S, Dahi P. Optimizing high dose melphalan. Blood Rev 2024; 64:101162. [PMID: 38097487 DOI: 10.1016/j.blre.2023.101162] [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: 09/21/2023] [Revised: 11/13/2023] [Accepted: 12/05/2023] [Indexed: 03/12/2024]
Abstract
Melphalan, has been a major component of myeloma therapy since the 1950s. In the context of hematopoietic cell transplantation (HCT), high dose melphalan (HDM) is the most common conditioning regimen used due to its potent anti-myeloma effects and manageable toxicities. Common toxicities associated with HDM include myelosuppression, gastrointestinal issues, and mucositis. Established approaches to reduce these toxicities encompass dose modification, nausea prophylaxis with 5HT3 receptor antagonists, cryotherapy, amifostine use, and growth factors. Optimization of melphalan exposure through personalized dosing and its combination with other agents like busulfan, or bendamustine show promise. Propylene glycol-free melphalan (Evomela) represents a novel formulation aiming to enhance drug stability and reduce adverse effects. This review explores strategies to enhance the efficacy and mitigate the toxicity of HDM in multiple myeloma. Future directions involve exploring these strategies in clinical trials to improve the safety and efficacy of HDM, thereby enhancing outcomes for multiple myeloma patients undergoing autologous HCT.
Collapse
Affiliation(s)
- Gunjan Shah
- Adult BMT Service Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, United States of America.
| | - Sergio Giralt
- Adult BMT Service Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, United States of America.
| | - Parastoo Dahi
- Adult BMT Service Memorial Sloan Kettering Cancer Center, 530 East 74th Street, New York, NY 10021, United States of America.
| |
Collapse
|
147
|
Davis JA, Dima D, Ahmed N, DeJarnette S, McGuirk J, Jia X, Raza S, Khouri J, Valent J, Anwer F, Abdallah AO, Hashmi H. Impact of Frailty on Outcomes after Chimeric Antigen Receptor T Cell Therapy for Patients with Relapsed/Refractory Multiple Myeloma. Transplant Cell Ther 2024; 30:298-305. [PMID: 38142943 DOI: 10.1016/j.jtct.2023.12.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/29/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
The literature is limited regarding outcomes in older adults and frail patients receiving BCMA-directed chimeric antigen receptor T cell therapy (CAR-T) for relapsed or refractory multiple myeloma. Here we describe the safety and efficacy of CAR-T in these clinically important subgroups treated in a real-world setting. Frailty was defined as a frail score ≥2 using the simplified frailty index (score based on age + Eastern Cooperative Oncology Group [ECOG] Performance Status + Hematopoietic Cell Transplantation Comorbidity Index [HCT-CI]). Of the 136 patients analyzed (age range, 41 to 81 years), 83 (61%) were considered frail at the time of CAR-T infusion. Compared to the nonfrail group, the frail group had higher proportions of patients with renal insufficiency (18% versus 6%), high-risk cytogenetics (45% versus 35%), extramedullary disease (51% versus 43%), and ECOG Performance Status ≥2 (18% versus 2%), and worse HCT-CI (3 versus 1). Although patients in the frail group had a higher incidence of immune effector cell-associated neurotoxicity syndrome (ICANS) (39% versus 17%), the incidences of all- grade cytokine release syndrome (CRS), as well as high-grade CRS and ICANS, were similar in the 2 groups. With a median follow-up of 7 months, the median progression-free survival was 6.9 months in the frail group versus 11.1 months in the nonfrail group (P = .028). The median overall survival was 14 months in the frail group and was not reached in the nonfrail group (P = .025). This study highlights the tolerable safety and reasonable efficacy of CAR-T for frail myeloma patients in a real-world practice. Although the frail patients did not experience any excessive high-grade toxicities, they did have inferior efficacy outcomes.
Collapse
Affiliation(s)
- James A Davis
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Department of Hematology-Oncology, Medical University of South Carolina, Charleston, South Carolina
| | - Danai Dima
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Center, Cleveland, Ohio
| | - Nausheen Ahmed
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas
| | - Shaun DeJarnette
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas
| | - Joseph McGuirk
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas
| | - Xuefei Jia
- Department of Biostatistics and Quantitative Health Science, Cleveland Clinic, Cleveland, Ohio
| | - Shahzad Raza
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Center, Cleveland, Ohio
| | - Jack Khouri
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Center, Cleveland, Ohio
| | - Jason Valent
- Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Center, Cleveland, Ohio
| | - Faiz Anwer
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Department of Hematology-Oncology, Cleveland Clinic, Taussig Cancer Center, Cleveland, Ohio
| | - Al-Ola Abdallah
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Westwood, Kansas
| | - Hamza Hashmi
- US Myeloma Innovations Research Collaborative, Kansas City, Kansas; Department of Hematology-Oncology, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
148
|
Liu Y, Ho C, Yu W, Huang Y, Miller J, Gao Q, Syed M, Ma Y, Wang M, Maciag L, Petrova-Drus K, Zhu M, Yao J, Vanderbilt C, Durham B, Benhamida J, Ewalt MD, Dogan A, Roshal M, Nafa K, Arcila ME. Quantification of Measurable Residual Disease Detection by Next-Generation Sequencing-Based Clonality Testing in B-Cell and Plasma Cell Neoplasms. J Mol Diagn 2024; 26:168-178. [PMID: 38103591 PMCID: PMC10918645 DOI: 10.1016/j.jmoldx.2023.11.009] [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: 07/04/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 12/19/2023] Open
Abstract
Next-generation sequencing (NGS)-based measurable residual disease (MRD) monitoring in post-treatment settings can be crucial for relapse risk stratification in patients with B-cell and plasma cell neoplasms. Prior studies have focused on validation of various technical aspects of the MRD assays, but more studies are warranted to establish the performance characteristics and enable standardization and broad utilization in routine clinical practice. Here, the authors describe an NGS-based IGH MRD quantification assay, incorporating a spike-in calibrator for monitoring B-cell and plasma cell neoplasms based on their unique IGH rearrangement status. Comparison of MRD status (positive or undetectable) by NGS and flow cytometry (FC) assays showed high concordance (91%, 471/519 cases) and overall good linear correlation in MRD quantitation, particularly for chronic lymphocytic leukemia and B-lymphoblastic leukemia/lymphoma (R = 0.85). Quantitative correlation was lower for plasma cell neoplasms, where underestimation by FC is a known limitation. No significant effects on sequencing efficiency by the spike-in calibrator were observed, with excellent inter- and intra-assay reproducibility within the authors' laboratory, and in comparison to an external laboratory, using the same assay and protocols. Assays performed both at internal and external laboratories showed highly concordant MRD detection (100%) and quantitation (R = 0.97). Overall, this NGS-based MRD assay showed highly reproducible results with quantitation that correlated well with FC MRD assessment, particularly for B-cell neoplasms.
Collapse
Affiliation(s)
- Ying Liu
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Caleb Ho
- Loxo Oncology, Inc., Stamford, Connecticut
| | - Wayne Yu
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ying Huang
- Invivoscribe, Inc., San Diego, California
| | | | - Qi Gao
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mustafa Syed
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuanyuan Ma
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meiyi Wang
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lidia Maciag
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kseniya Petrova-Drus
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Menglei Zhu
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - JinJuan Yao
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad Vanderbilt
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benjamin Durham
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jamal Benhamida
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark D Ewalt
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Dogan
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Khedoudja Nafa
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Diagnostic Molecular Pathology, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Hematopathology Service, Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
149
|
Scheid C, Landgren O. Early detection of multiple myeloma: A first step towards a screening programme. Br J Haematol 2024; 204:393-394. [PMID: 38168005 DOI: 10.1111/bjh.19274] [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: 12/06/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024]
Abstract
Novel therapies for multiple myeloma have substantially improved the prognosis, but before patients are able to benefit from treatment, a diagnosis must have been made. The Myeloma UK laboratory working group has developed a three-part tool box to help primary care physicians to suspect myeloma despite mostly vague symptoms, to do the right tests and to derive the proper conclusions from the results. Commentary on: Drayson et al. Laboratory practice is central to earlier myeloma diagnosis; utilising a primary care diagnostic tool and laboratory guidelines integrated into haematology services. Br J Haematol 2024;204:476-486.
Collapse
Affiliation(s)
- Christof Scheid
- University Hospital and Center for Integrated Oncology Cologne, Cologne, Germany
| | - Ola Landgren
- Division of Myeloma, Sylvester Comprehensive Cancer Center, University of Miami, Coral Gables, Florida, USA
| |
Collapse
|
150
|
Gibson CJ, Lindsley RC, Gondek LP. Clonal hematopoiesis in the setting of hematopoietic cell transplantation. Semin Hematol 2024; 61:9-15. [PMID: 38429201 PMCID: PMC10978245 DOI: 10.1053/j.seminhematol.2024.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 03/03/2024]
Abstract
Clonal hematopoiesis (CH) in autologous transplant recipients and allogeneic transplant donors has genetic features and clinical associations that are distinct from each other and from non-cancer populations. CH in the setting of autologous transplant is enriched for mutations in DNA damage response pathway genes and is associated with adverse outcomes, including an increased risk of therapy-related myeloid neoplasm and inferior overall survival. Studies of CH in allogeneic transplant donors have yielded conflicting results but have generally shown evidence of potentiated alloimmunity in recipients, with some studies showing an association with favorable recipient outcomes.
Collapse
Affiliation(s)
| | - R Coleman Lindsley
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
| | - Lukasz P Gondek
- Division of Hematologic Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD.
| |
Collapse
|