251
|
Thurlapati A, Roubal K, Davis JA, Shah SZ, Smith D, McGann M, Gaffney K, Cendagorta A, Maldonado A, Weeda E, Hashmi H. Stem Cell Mobilization for Multiple Myeloma Patients Receiving Daratumumab-Based Induction Therapy: A Real- World Experience. Transplant Cell Ther 2023; 29:340.e1-340.e4. [PMID: 36804934 DOI: 10.1016/j.jtct.2023.02.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/16/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023]
Abstract
For patients with newly diagnosed multiple myeloma (MM) undergoing high-dose chemotherapy and autologous stem cell transplantation (HDT-ASCT), hematopoietic stem cell mobilization can be affected by induction chemotherapy. In clinical trials, the addition of daratumumab (dara) to a triplet backbone lowered hematopoietic stem cell yield, necessitating the administration of plerixafor to achieve the desired yield for ASCT. Here we describe our experience of stem cell mobilization and collection after dara-based and non-dara-based induction regimens. This single-center retrospective analysis included patients with newly diagnosed MM who had received induction chemotherapy and were candidates for upfront HDT-ASCT. Based on the induction regimen used, patients were divided into 2 groups, RVd (lenalidomide, bortezomib, and dexamethasone) and DRVd (RVd with the addition of dara). Based on our institutional practice, patients received pegylated growth colony-stimulating factor (G-CSF) on day -3 (at 0900 hours) in combination with plerixafor on day -1 (at 2300 hours) as a preemptive mobilization strategy. Patients continued apheresis for 1 to 3 days until the goal dose of hematopoietic stem cells was collected (2.5 × 106 cells/kg for one ASCT and 5.0 × 106 cells/kg for 2 ASCTs). Patients with a suboptimal stem cell yield on day 1 received additional doses of plerixafor with or without G-CSF. A total of 101 patients with newly diagnosed MM who underwent mobilization between July 2021 and June 2022 were analyzed. The median patient age was 61 years (range, 36 to 80 years), and 51.5% of the cohort was female. Patients received a median of 5 (range, 2 to 12) cycles of induction chemotherapy, with a median of 4 (range, 2 to 12) cycles of DRVd and 6 (range, 3 to 12) cycles of RVd. The median number of CD34+ cells collected in the DRVd and the RVd groups was 6.54 × 106/kg and 6.78 × 106/kg, respectively. Target CD34+ stem cells were collected in a median of 1 day (range, 1 to 4 day) in each group. On average, more patients in the DRVd group compared to the RVd group received additional doses of plerixafor (51% versus 43%) and additional doses of GCSF (19% versus 14%) to achieve the target stem cell yield. There were no mobilization failures or grade 3+ mobilization-related adverse events reported in either group. The addition of daratumumab to the RVd induction regimen did not lead to any clinically significant differences in stem cell yield or number of collection days, provided that the patient received preemptive G-CSF and plerixafor. Patients with suboptimal collection on day 1 were able to collect adequate stem cells with additional doses of plerixafor with or without G-CSF.
Collapse
Affiliation(s)
- Aswani Thurlapati
- Department of Hematology and Bone Marrow Transplant, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Kiera Roubal
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - James A Davis
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Syed Z Shah
- Department of Hospital Medicine, Medical University of South Carolina, Columbia, South Carolina
| | - Deidra Smith
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Mary McGann
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Kelly Gaffney
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Alyssa Cendagorta
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Andy Maldonado
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Erin Weeda
- Department of Pharmacy, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | - Hamza Hashmi
- Department of Hematology and Bone Marrow Transplant, Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina.
| |
Collapse
|
252
|
Minnie SA, Waltner OG, Ensbey KS, Olver SD, Collinge AD, Sester DP, Schmidt CR, Legg SR, Takahashi S, Nemychenkov NS, Sekiguchi T, Driessens G, Zhang P, Koyama M, Spencer A, Holmberg LA, Furlan SN, Varelias A, Hill GR. TIGIT inhibition and lenalidomide synergistically promote antimyeloma immune responses after stem cell transplantation in mice. J Clin Invest 2023; 133:e157907. [PMID: 36512425 PMCID: PMC9927935 DOI: 10.1172/jci157907] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Autologous stem cell transplantation (ASCT) with subsequent lenalidomide maintenance is standard consolidation therapy for multiple myeloma, and a subset of patients achieve durable progression-free survival that is suggestive of long-term immune control. Nonetheless, most patients ultimately relapse, suggesting immune escape. TIGIT appears to be a potent inhibitor of myeloma-specific immunity and represents a promising new checkpoint target. Here we demonstrate high expression of TIGIT on activated CD8+ T cells in mobilized peripheral blood stem cell grafts from patients with myeloma. To guide clinical application of TIGIT inhibition, we evaluated identical anti-TIGIT antibodies that do or do not engage FcγR and demonstrated that anti-TIGIT activity is dependent on FcγR binding. We subsequently used CRBN mice to investigate the efficacy of anti-TIGIT in combination with lenalidomide maintenance after transplantation. Notably, the combination of anti-TIGIT with lenalidomide provided synergistic, CD8+ T cell-dependent, antimyeloma efficacy. Analysis of bone marrow (BM) CD8+ T cells demonstrated that combination therapy suppressed T cell exhaustion, enhanced effector function, and expanded central memory subsets. Importantly, these immune phenotypes were specific to the BM tumor microenvironment. Collectively, these data provide a logical rationale for combining TIGIT inhibition with immunomodulatory drugs to prevent myeloma progression after ASCT.
Collapse
Affiliation(s)
- Simone A. Minnie
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Olivia G. Waltner
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kathleen S. Ensbey
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Stuart D. Olver
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Alika D. Collinge
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David P. Sester
- Translational Research Institute, Woolloongabba, Queensland, Australia
- Hugh Green Cytometry Centre, Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Christine R. Schmidt
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Samuel R.W. Legg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Shuichiro Takahashi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Tomoko Sekiguchi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Ping Zhang
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Andrew Spencer
- Australian Center for Blood Diseases, Monash University and
- Malignant Haematology and Stem Cell Transplantation, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Clinical Haematology, Monash University, Melbourne, Victoria, Australia
| | - Leona A. Holmberg
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology and
| | - Scott N. Furlan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, St. Lucia, Queensland, Australia
| | - Geoffrey R. Hill
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Medical Oncology and
| |
Collapse
|
253
|
Bhatt P, Kloock C, Comenzo R. Relapsed/Refractory Multiple Myeloma: A Review of Available Therapies and Clinical Scenarios Encountered in Myeloma Relapse. Curr Oncol 2023; 30:2322-2347. [PMID: 36826140 PMCID: PMC9954856 DOI: 10.3390/curroncol30020179] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Multiple myeloma remains an incurable disease with the usual disease course requiring induction therapy, autologous stem cell transplantation for eligible patients, and long-term maintenance. Risk stratification tools and cytogenetic alterations help inform individualized therapeutic choices for patients in hopes of achieving long-term remissions with preserved quality of life. Unfortunately, relapses occur at different stages of the course of the disease owing to the biological heterogeneity of the disease. Addressing relapse can be complex and challenging as there are both therapy- and patient-related factors to consider. In this broad scoping review of available therapies in relapsed/refractory multiple myeloma (RRMM), we cover the pharmacologic mechanisms underlying active therapies such as immunomodulatory agents (IMiDs), proteasome inhibitors (PIs), monoclonal antibodies (mAbs), traditional chemotherapy, and Venetoclax. We then review the clinical data supporting the use of these therapies, organized based on drug resistance/refractoriness, and the role of autologous stem cell transplant (ASCT). Approaches to special situations during relapse such as renal impairment and extramedullary disease are also covered. Lastly, we look towards the future by briefly reviewing the clinical data supporting the use of chimeric antigen receptor (CAR-T) therapy, bispecific T cell engagers (BITE), and Cereblon E3 Ligase Modulators (CELMoDs).
Collapse
Affiliation(s)
- Parva Bhatt
- Correspondence: (P.B.); (R.C.); Tel.: +1-617-636-6454
| | | | | |
Collapse
|
254
|
Bici A, Pianko MJ, Nachar VR. Incidence and risk factors for bacterial infection using bortezomib, lenalidomide, and dexamethasone (RVd) in newly diagnosed multiple myeloma. Leuk Lymphoma 2023; 64:407-414. [PMID: 36308285 PMCID: PMC9993956 DOI: 10.1080/10428194.2022.2138380] [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: 06/24/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 02/17/2023]
Abstract
Infections are an important cause of morbidity and mortality in newly diagnosed multiple myeloma (NDMM), but the real-world risk using modern induction regimens such as bortezomib, lenalidomide, and dexamethasone (RVd) is not well described. We performed a retrospective single-center cohort study to identify infections and risk factors in patients treated with first-line RVd from January 2014 to January 2020 and collected demographic and clinical data. Of 144 patients treated with RVd for NDMM, 21 patients (14.5%) experienced a bacterial infection during induction, of which 8 (5.5%) were grade 3 infections despite a low rate of antibiotic prophylaxis use (12%). Grade 3 neutropenia occurred in 11% of patients, 2% had febrile neutropenia and there were no deaths from infection. On multivariable analysis, age, smoking history, diabetes, antibiotic use in the 60 days preceding the start of RVd, and high-risk cytogenetics were associated with higher risk of bacterial infection.
Collapse
Affiliation(s)
- Anisa Bici
- Department of Pharmacy Services, Michigan Medicine and the University of Michigan College of Pharmacy, Ann Arbor, MI, United States
| | - Matthew J. Pianko
- Division of Hematology and Oncology, Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Victoria R Nachar
- Department of Pharmacy Services, University of Michigan Rogel Cancer Center, Ann Arbor, MI, United States
| |
Collapse
|
255
|
Yong K, Wilson W, de Tute RM, Camilleri M, Ramasamy K, Streetly M, Sive J, Bygrave CA, Benjamin R, Chapman M, Chavda SJ, Phillips EH, Del Mar Cuadrado M, Pang G, Jenner R, Dadaga T, Kamora S, Cavenagh J, Clifton-Hadley L, Owen RG, Popat R. Upfront autologous haematopoietic stem-cell transplantation versus carfilzomib-cyclophosphamide-dexamethasone consolidation with carfilzomib maintenance in patients with newly diagnosed multiple myeloma in England and Wales (CARDAMON): a randomised, phase 2, non-inferiority trial. Lancet Haematol 2023; 10:e93-e106. [PMID: 36529145 DOI: 10.1016/s2352-3026(22)00350-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/16/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Standard-of-care treatment for patients with newly diagnosed multiple myeloma is bortezomib-based induction followed by high-dose melphalan and autologous haematopoietic stem-cell transplantation (HSCT) and lenalidomide maintenance. We aimed to evaluate whether an immunomodulatory-free carfilzomib-based induction, consolidation, and maintenance protocol without autologous HSCT was non-inferior to the same induction regimen followed by autologous HSCT and maintenance. METHODS CARDAMON is a randomised, open-label, phase 2 trial in 19 hospitals in England and Wales, UK. Newly diagnosed, transplantation-eligible patients with multiple myeloma aged 18 years or older with an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 received four 28-day cycles of carfilzomib (56 mg/m2 intravenously on days 1, 2, 8, 9, 15, and 16), cyclophosphamide (500 mg orally on days 1, 8, and 15), and dexamethasone (40 mg orally on days 1, 8, 15, and 22; KCd), followed by peripheral blood stem cell mobilisation. Patients with at least a partial response were randomly assigned (1:1) to either high-dose melphalan and autologous HSCT or four cycles of KCd. All randomised patients received 18 cycles of carfilzomib maintenance (56 mg/m2 intravenously on days 1, 8, and 15). The primary outcomes were the proportion of patients with at least a very good partial response after induction and difference in progression-free survival rate at 2 years from randomisation (non-inferiority margin 10%), both assessed by intention to treat. Safety was assessed in all patients who started treatment. The trial is registered with ClinicalTrials.gov (NCT02315716); recruitment is complete and all patients are in follow-up. FINDINGS Between June 16, 2015, and July 8, 2019, 281 patients were enrolled, with 218 proceeding to randomisation (109 assigned to the KCd consolidation group [99 of whom completed consolidation] and 109 to the HSCT group [104 of whom underwent transplantation]). A further seven patients withdrew before initiation of carfilzomib maintenance (two in the KCd consolidation group vs five in the HSCT group). Median age was 59 years (IQR 52 to 64); 166 (59%) of 281 patients were male and 115 (41%) were female. 152 (71%) of 214 patients with known ethnicity were White, 37 (17%) were Black, 18 (8%) were Asian, 5 (2%) identified as Mixed, and 2 (1%) identified as other. Median follow-up from randomisation was 40·2 months (IQR 32·7 to 51·8). After induction, 162 (57·7%; 95% CI 51·6 to 63·5) of 281 patients had at least a very good partial response. The 2-year progression-free survival was 75% (95% CI 65 to 82) in the HSCT group versus 68% (95% CI 58 to 76) in the KCd group (difference -7·2%, 70% CI -11·1 to -2·8), exceeding the non-inferiority margin. The most common grade 3-4 events during KCd induction and consolidation were lymphocytopenia (72 [26%] of 278 patients who started induction; 15 [14%] of 109 patients who started consolidation) and infection (50 [18%] of 278 for induction; 15 [14%] of 109 for consolidation), and during carfilzomib maintenance were hypertension (20 [21%] of 97 patients in the KCd consolidation group vs 23 [23%] of 99 patients in the HSCT group) and infection (16 [16%] of 97 patients vs 25 [25%] of 99). Treatment-related serious adverse events at any point during the trial were reported in 109 (39%) of 278 patients who started induction, with infections (80 [29%]) being the most common. Treatment-emergent deaths were reported in five (2%) of 278 patients during induction (three from infection, one from cardiac event, and one from renal failure) and one of 99 patients during maintenance after autologous HSCT (oesophageal carcinoma). INTERPRETATION KCd did not meet the criteria for non-inferiority compared with autologous HSCT, but the marginal difference in progression-free survival suggests that further studies are warranted to explore deferred autologous HSCT in some subgroups, such as individuals who are MRD negative after induction. FUNDING Cancer Research UK and Amgen.
Collapse
Affiliation(s)
- Kwee Yong
- Cancer Institute, University College London, London, UK; Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK.
| | - William Wilson
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Ruth M de Tute
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Marquita Camilleri
- Cancer Institute, University College London, London, UK; Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Karthik Ramasamy
- Department of Clinical Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Matthew Streetly
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jonathan Sive
- Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Ceri A Bygrave
- Department of Haematology, University Hospital of Wales, Cardiff, UK
| | | | - Michael Chapman
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | | | | | | | - Gavin Pang
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Richard Jenner
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Tushhar Dadaga
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Sumaiya Kamora
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - James Cavenagh
- Department of Haemato-Oncology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Laura Clifton-Hadley
- Cancer Research UK and UCL Cancer Trials Centre, University College London, London, UK
| | - Roger G Owen
- Haematological Malignancy Diagnostic Service, St James's University Hospital, Leeds, UK
| | - Rakesh Popat
- Clinical Haematology Department, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
256
|
Urushihara R, Takezako N, Yoroidaka T, Yamashita T, Murata R, Satou K, Nakao S, Takamatsu H. Eight-color multiparameter flow cytometry (EuroFlow-NGF) is as sensitive as next-generation sequencing in detecting minimal/measurable residual disease in autografts of patients with multiple myeloma. EJHAEM 2023; 4:184-191. [PMID: 36819171 PMCID: PMC9928796 DOI: 10.1002/jha2.633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 01/25/2023]
Abstract
The prognostic value of minimal/measurable residual disease (MRD) detection in autografts of patients with multiple myeloma (MM) in an autologous stem-cell transplantation setting has been reported. Next-generation flow (NGF) cytometry has lower sensitivity (2 × 10-6) to detect MRD than next-generation sequencing (NGS) (<10-6). We compared the clinical value of high-sensitivity NGF (cutoff: <10-6) and NGS (cutoff: 10-6) for the detection of MRD in the cryopreserved autografts of 49 patients with newly diagnosed MM. The sensitivity test using frozen/thawed autografts revealed a strong correlation among MRD levels of 5 × 10-7 and 1 × 10-4 (r = 0.9997, p < 0.0001) when an adequate number of cells were analyzed. Autograft MRD levels determined using NGF and NGS were highly correlated (r = 0.811, p < 0.0001). MRD-negative patients identified with NGF (cutoff: <10-6) showed significantly longer progression-free survival (PFS) than MRD-positive patients (p = 0.026). The PFS of MRD-negative patients determined by NGS (cutoff: 10-6) was similar to that determined by NGF. These results show that the high-sensitivity NGF method can assess MRD in frozen/thawed autografts, and its prognostic value is comparable to that of NGS.
Collapse
Affiliation(s)
- Ryota Urushihara
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | - Naoki Takezako
- Department of HematologyDisaster Medical Center of JapanTachikawaJapan
| | - Takeshi Yoroidaka
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | | | - Ryoichi Murata
- Division of Internal MedicineKeiju Kanazawa HospitalKanazawaJapan
| | - Kenji Satou
- Faculty of Transdisciplinary Sciences for InnovationInstitute of Transdisciplinary Sciences for InnovationKanazawa UniversityKanazawaJapan
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | - Hiroyuki Takamatsu
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
- Faculty of Transdisciplinary Sciences for InnovationInstitute of Transdisciplinary Sciences for InnovationKanazawa UniversityKanazawaJapan
| |
Collapse
|
257
|
Beksac M, Hayden P. Upfront autologous transplantation still improving outcomes in patients with multiple myeloma. Lancet Haematol 2023; 10:e80-e82. [PMID: 36529146 DOI: 10.1016/s2352-3026(22)00360-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Meral Beksac
- Department of Hematology, Cebeci Hospital, Ankara University, Ankara 06620, Türkiye.
| | - Patrick Hayden
- Department of Haematology, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| |
Collapse
|
258
|
Engelhardt M, Wäsch R. Carfilzomib, lenalidomide, and dexamethasone maintenance for multiple myeloma: when and for whom? Lancet Oncol 2023; 24:118-119. [PMID: 36642082 DOI: 10.1016/s1470-2045(22)00792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023]
Affiliation(s)
- Monika Engelhardt
- Department of Hematology and Oncology, Interdisciplinary Cancer Center, University of Freiburg, 79106 Freiburg, Germany.
| | - Ralph Wäsch
- Department of Hematology and Oncology, Interdisciplinary Cancer Center, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
259
|
Mizuguchi M, Okamoto Y, Yagi H, Kagawa K, Sekimoto E, Shibata H, Shigekiyo T, Ozaki S. Clinical relevance of high-risk cytogenetic abnormalities and the second revision of the International Staging System (R2-ISS) in patients with multiple myeloma in clinical practice. Int J Hematol 2023; 117:718-728. [PMID: 36692689 DOI: 10.1007/s12185-023-03541-x] [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: 11/08/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
High-risk cytogenetic abnormalities (HRCAs) are the most critical factor affecting prognosis in multiple myeloma (MM). However, the clinical significance of HRCAs in routine practice has not been fully elucidated. We retrospectively analyzed clinical features and outcome in 60 newly diagnosed MM patients with or without HRCAs including t(4;14), t(14;16), del(17p), and 1q gain/amplification. The median age was 71 years (range, 35-90). Abnormalities with t(4;14), t(14;16), del(17p), and 1q gain/amplification were found in 10, 1, 6, and 21/14 patients, respectively, and 10 patients had ≥ 2 HRCAs. Patients with HRCAs exhibited progressive clinical features such as anemia, high β2-microglobulin, and high LDH. Symptomatic relapse was more common in patients with HRCAs. The median progression-free survival (PFS) by number of HRCAs (0, 1, and ≥ 2) was 51.7, 21.4, and 26.1 months (p = 0.011), and the median overall survival (OS) was not reached, 60.7, and 46.8 months (p = 0.045), respectively. Multivariate analysis revealed that HRCAs were an independent factor for PFS. Accordingly, the second revision of International Staging System (R2-ISS), which incorporates HRCA scores, was more useful for prognostic stratification (p = 0.0023). These results suggest that presence of multiple HRCAs including 1q gain/amplification is associated with advanced stage and poor prognosis in clinical practice as well.
Collapse
Affiliation(s)
- Makiko Mizuguchi
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Yasunobu Okamoto
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Hikaru Yagi
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Kumiko Kagawa
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Etsuko Sekimoto
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan.,Department of Internal Medicine, Tenma Hospital, 1-5-1 Kuramoto, Tokushima, 770-0042, Japan
| | - Hironobu Shibata
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Toshio Shigekiyo
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Shuji Ozaki
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan.
| |
Collapse
|
260
|
Safety and efficacy of autologous stem cell transplantation in dialysis-dependent myeloma patients-The DIADEM study from the chronic malignancies working party of the EBMT. Bone Marrow Transplant 2023; 58:424-429. [PMID: 36681775 DOI: 10.1038/s41409-023-01915-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/27/2022] [Accepted: 01/10/2023] [Indexed: 01/22/2023]
Abstract
The role of high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) in the treatment of myeloma (MM) patients with severe and/or dialysis-dependent renal impairment remains uncertain. We report on the outcomes of 110 patients (median age 57 years) who had become dialysis-dependent pre-ASCT and who underwent a first ASCT between 1997 and 2017. Sixty-three (57%) patients had light chain MM. All patients required dialysis (94% hemodialysis and 6% peritoneal). Forty-four of 71 (62%) patients received bortezomib-based induction regimens and 42 (39%) patients had achieved at least a very good partial response (VGPR) pre-ASCT. Melphalan dosing was as follows: ≤140 mg/m2 (82%), and >140 mg/m2 (18%). The median PFS after ASCT was 35 months (95% CI: 21.5-42.2) and the median OS 102 months (95% CI: 70.4-129.1). At 1, 2, and 5 years after ASCT, 8% (95% CI 3-14%), 13% (6-20%), and 20% (12-29%) of patients, respectively, had achieved dialysis independence. In multivariate analyses of OS and PFS including age at ASCT, response at ASCT, and year of ASCT, younger age at ASCT and better response at ASCT (CR/VGPR/PR vs. MR/SD/progression) were significantly associated with better OS and PFS.
Collapse
|
261
|
Parikh RH, Lonial S. Chimeric antigen receptor T-cell therapy in multiple myeloma: A comprehensive review of current data and implications for clinical practice. CA Cancer J Clin 2023; 73:275-285. [PMID: 36627265 DOI: 10.3322/caac.21771] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/10/2022] [Accepted: 12/02/2022] [Indexed: 01/12/2023] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy defined by the clonal proliferation of transformed plasma cells. Despite tremendous advances in the treatment paradigm of MM, a cure remains elusive for most patients. Although long-term disease control can be achieved in a very large number of patients, the acquisition of tumor resistance leads to disease relapse, especially in patients with triple-class refractory MM (defined as resistance to immunomodulatory agents, proteosome inhibitors, and monoclonal antibodies). There is an unmet need for effective treatment options in these patients. Chimeric antigen receptor (CAR) T-cell therapy is a novel approach that has demonstrated promising efficacy in the treatment of relapsed, refractory MM (RRMM). These genetically modified cellular therapies have demonstrated deep and durable remissions in other B-cell malignancies, and current efforts aim to achieve similar results in patients with RRMM. Early studies have demonstrated remarkable response rates with CAR T-cell therapy in RRMM; however, durable responses with CAR T-cell therapies in myeloma have yet to be realized. In this comprehensive review, the authors describe the development of CAR T-cell therapies in myeloma, the outcomes of notable clinical trials, the toxicities and limitations of CAR T-cell therapies, and the strategies to overcome therapeutic challenges of CAR T cells in the hope of achieving a cure for multiple myeloma.
Collapse
Affiliation(s)
- Rujul H Parikh
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
262
|
Carfilzomib induction, consolidation, and maintenance with or without autologous stem-cell transplantation in patients with newly diagnosed multiple myeloma: pre-planned cytogenetic subgroup analysis of the randomised, phase 2 FORTE trial. Lancet Oncol 2023; 24:64-76. [PMID: 36528035 DOI: 10.1016/s1470-2045(22)00693-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 10/21/2022] [Accepted: 11/03/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with newly diagnosed multiple myeloma and high-risk cytogenetic abnormalities (HRCA) represent an unmet medical need. In the FORTE trial, lenalidomide and dexamethasone plus carfilzomib (KRd) induction resulted in a higher proportion of patients with at least a very good partial response as compared with carfilzomib, cyclophosphamide, and dexamethasone (KCd), and carfilzomib plus lenalidomide maintenance prolonged progression-free survival compared with lenalidomide maintenance. In this prespecified analysis of the FORTE trial, we described the outcomes of enrolled patients according to their cytogenetic risk. METHODS The UNITO-MM-01/FORTE was a randomised, open-label, phase 2 trial done at 42 Italian academic and community practice centres, which enrolled transplant-eligible patients with newly diagnosed multiple myeloma aged 18-65 years. Eligible patients had newly diagnosed multiple myeloma based on standard International Myeloma Working Group criteria, a Karnofsky performance status of at least 60%, and had not received any previous treatment with anti-myeloma therapy. At enrolment, patients were stratified according to International Staging System stage (I vs II/III) and age (<60 years vs 60-65 years) and randomly assigned (1:1:1) to KRd plus autologous stem-cell transplantation (ASCT; four 28-day induction cycles with KRd, melphalan at 200 mg/m2 and ASCT [MEL200-ASCT], followed by four 28-day KRd consolidation cycles), 12 28-day KRd cycles, or KCd plus ASCT (four 28-day induction cycles with KCd, MEL200-ASCT, and four 28-day KCd consolidation cycles), using a web-based system (block randomisation, block size of 12). Carfilzomib was administered at 20 mg/m2 on days 1 and 2 of cycle 1, followed by 36 mg/m2 intravenously administered on days 8, 9, 15, and 16 of cycle 1, and then 36 mg/m2 intravenously administered for all subsequent doses on days 1, 2, 8, 9, 15, 16; lenalidomide 25 mg was administered orally on days 1-21; cyclophosphamide 300 mg/m2 was administered orally on days 1, 8, and 15; and dexamethasone 20 mg was administered orally or intravenously on days 1, 2, 8, 9, 15, 16, 22, and 23. After the consolidation phase, patients were stratified according to induction-consolidation treatment and randomly assigned (1:1; block size of 8) to maintenance treatment with carfilzomib plus lenalidomide or lenalidomide alone. Carfilzomib 36 mg/m2 was administered intravenously on days 1-2 and days 15-16, every 28 days for up to 2 years, and lenalidomide 10 mg was administered orally on days 1-21 every 28 days until progression or intolerance in both groups. The primary endpoints were the proportion of patients with at least a very good partial response after induction with KRd versus KCd and progression-free survival with carfilzomib plus lenalidomide versus lenalidomide alone as maintenance treatment. In this preplanned analysis, we included patients enrolled in the FORTE trial with complete cytogenetic data on del(17p), t(4;14), t(14;16), del(1p), gain(1q) (3 copies), and amp(1q) (≥4 copies) assessed by fluorescence in-situ hybridisation analysis on CD138-positive sorted cells. We assessed progression-free survival, overall survival, minimal residual disease negativity, and 1-year sustained minimal residual disease negativity according to the presence of zero, one, and two or more HRCA across treatment groups. The FORTE trial is ongoing, and registered with ClinicalTrials.gov, NCT02203643. FINDINGS Between Feb 23, 2015, and April 5, 2017, 477 patients were enrolled, of whom 396 (83%) had complete cytogenetic data and were analysed (176 [44%] of whom were women and 220 [56%] were men). The median follow-up from first randomisation was 51 months (IQR 46-56). 4-year progression-free survival was 71% (95% CI 64-78) in patients with zero HRCA, 60% (95% CI 52-69) in patients with one HRCA, and 39% (95% CI 30-50) in patients with two or more HRCA. Compared with patients with zero HRCA, the risk of progression or death was similar in patients with one HRCA (hazard ratio [HR] 1·33 [95% CI 0·90-1·97]; p=0·15) and higher in patients with two or more HRCA (HR 2·56 [95% CI 1·74-3·75]); p<0·0001) across the induction-intensification-consolidation groups. Moreover, the risk of progression or death was also higher in patients with two or more HRCA versus those with one HRCA (HR 1·92 [95% CI 1·34-2·76]; p=0·0004). 4-year overall survival from the first randomisation was 94% (95% CI 91-98) in patients with zero HRCA, 83% (95% CI 76-90) in patients with one HRCA, and 63% (95% CI 54-74) in patients with two or more HRCA. Compared with patients with zero HRCA, the risk of death was significantly higher in patients with one HRCA (HR 2·55 [95% CI 1·22-5·36]; p=0·013) and two or more HRCA (HR 6·53 [95% CI 3·24-13·18]; p<0·0001). Patients with two or more HRCA also had a significantly higher risk of death than those with one HRCA (HR 2·56 [95% CI 1·53-4·28]; p=0·0004). The rates of 1-year sustained minimal residual disease negativity were similar in patients with zero HRCA (53 [35%] of 153] and with one HRCA (57 [41%] of 138) and were lower in patients with two or more HRCA (25 [24%] of 105). The median duration of follow-up from second randomisation was 37 months (IQR 33-42). 3-year progression-free survival from the second randomisation was 80% (95% CI 74-88) in patients with zero HRCA, 68% (95% CI 59-78) in patients with one HRCA, and 53% (95% CI 42-67) in patients with two or more HRCA. The risk of progression or death was higher in patients with one HRCA (HR 1·68 [95% CI 1·01-2·80]; p=0·048) and two or more HRCA (2·74 [95% CI 1·60-4·69], p=0·0003) than in patients with zero HRCA. INTERPRETATION This preplanned analysis of the FORTE trial showed that carfilzomib-based induction-intensification-consolidation regimens are effective strategies in patients with standard risk (zero HRCA) and high-risk (one HRCA) myeloma, resulting in similar rates of progression-free survival and 1-year sustained minimal residual disease negativity. Despite promising progression-free survival, patients with ultra-high-risk disease (those with 2 or more HRCA) still have an increased risk of progression and death and therefore represent an unmet medical need. FUNDING Amgen and Celgene/Bristol Myers Squibb.
Collapse
|
263
|
Tanenbaum B, Miett T, Patel SA. The emerging therapeutic landscape of relapsed/refractory multiple myeloma. Ann Hematol 2023; 102:1-11. [PMID: 36462062 PMCID: PMC10888499 DOI: 10.1007/s00277-022-05058-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/13/2022] [Indexed: 12/07/2022]
Abstract
From a historic lens, treatment for patients with relapsed/refractory multiple myeloma (R/R MM) has advanced significantly since the advent of immunomodulatory agents (IMiDs) in the 1990s, proteasome inhibitors in the 2000s, monoclonal antibodies in the 2010s, and CAR-T treatments in the 2020s. However, the availability of multiple new therapies has also created significant ambiguity regarding therapy selection and sequencing, as consensus guidelines are limited, and cross-trial comparisons of the novel agents are challenging. In this focused review, we discuss the novel Food & Drug Administration (FDA)-approved medications for R/R MM, including the recently approved first-in-class BCMA-directed bispecific antibody teclistamab. We highlight the seminal clinical trials data and discuss optimal sequencing considerations based on the goal of treatment, with an emphasis on the two novel CAR-T cell products. We consider the limited tolerability of certain agents, prospects for our aging population, and financial aspects of these therapies. Finally, we spotlight ongoing trials involving promising agents making their way through the pharmacologic pipeline including the BCMA-directed bispecific antibody elranatamab and the GPRC5D-directed bispecific antibody talquetamab. We summarize our recommendations based on the best available evidence as we enter 2023.
Collapse
Affiliation(s)
- Benjamin Tanenbaum
- Department of Medicine, UMass Chan Medical School, UMass Memorial Medical Center, 55 Lake Ave. North, Worcester, MA, 01655, USA
| | - Timothy Miett
- Department of Medicine, UMass Chan Medical School, UMass Memorial Medical Center, 55 Lake Ave. North, Worcester, MA, 01655, USA
| | - Shyam A Patel
- Department of Medicine - Division of Hematology/Oncology, UMass Chan Medical School, UMass Memorial Medical Center, 55 Lake Ave. North, Worcester, MA, 01655, USA.
- Center for Clinical and Translational Science (CCTS), UMass Chan Medical School, 55 Lake Ave. North, Worcester, MA, 01655, USA.
| |
Collapse
|
264
|
Garrido D, Bove V, Villano F, Riva E. Survival Analysis of Newly Diagnosed Multiple Myeloma Patients after Frontline Autologous Stem Cell Transplantation in a Real-Life Setting. ACTA MEDICA (HRADEC KRALOVE) 2023; 66:117-121. [PMID: 38511422 DOI: 10.14712/18059694.2024.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Autologous stem cell transplantation (ASCT) is the standard consolidation option for transplant-eligible patients with multiple myeloma (MM). The aim of this study is to report the overall survival (OS) and progression-free survival (PFS) outcomes after frontline ASCT in newly-diagnosed MM (NDMM) patients in a real-world setting. METHODS We conducted a retrospective, survival analysis of all NDMM patients included in the MM Uruguayan Registry. RESULTS We included 151 NDMM patients treated with induction therapy followed by high-dose melphalan and ASCT as consolidation. The median age at diagnosis was 59 years, and the international staging system (ISS) risk groups were ISS-III 32.9%, ISS-II 37.8%, and ISS-I 29.4%. Frontline induction regimens included bortezomib in 61.6% of cases, and maintenance therapy was used in 63.9% of reported cases. With a median follow-up of 42 months, the 36-month OS and PFS for the whole group were 82.4% (95% CI 75.9% to 89.4%) and 63.8% (95% CI 55.6% to 73.3%), respectively, median OS of 98 months and median PFS of 47 months. The 100-month OS and PFS for the entire group were 48.0% (95% CI 34.9% to 66.0%) and 17.3% (95% CI 8.4% to 35.8%), respectively. CONCLUSION ASCT is a feasible, safe, and potent strategy that provides a prolonged median OS and PFS in NDMM patients. This approach can be implemented in low-income countries.
Collapse
Affiliation(s)
- David Garrido
- Cátedra de Hematología, Hospital de Clínicas, Av Italia, sn; Montevideo, CP 11600, Uruguay
| | - Virginia Bove
- Hospital Central de las Fuerzas Armadas, Ocho de octubre 3020, CP 11600, Montevideo, Uruguay
| | | | - Eloísa Riva
- Cátedra de Hematología, Hospital de Clínicas, Av Italia, sn; Montevideo, CP 11600, Uruguay.
| |
Collapse
|
265
|
Mikulski D, Kościelny K, Nowicki M, Wawrzyniak E, Kalwas M, Kowalik M, Pryt M, Sęczkowska E, Świątek A, Wierzbowska A, Fendler W. Neutrophil to lymphocyte ratio (NLR) impact on the progression-free survival and overall survival of multiple myeloma patients treated with high-dose chemotherapy and autologous stem cell transplantation. Leuk Lymphoma 2023; 64:98-106. [PMID: 36318865 DOI: 10.1080/10428194.2022.2136946] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
High-dose chemotherapy with autologous stem-cell transplantation (ASCT) remains the standard of care in multiple myeloma (MM) patients. This retrospective study aimed to assess the impact of neutrophil-to-lymphocyte ratio (NLR) and other complete blood count (CBC)-based predictors on PFS and OS of transplant-eligible MM patients. The CBC-based biomarkers were evaluated in a single-center cohort of 176 MM patients at three time points: at the diagnosis, the time of ASCT, and +100 d after ASCT. Univariable and multivariable Cox's regression analyses and Kaplan-Meier estimate were used in statistical analysis. NLR at ASCT (HR 1.15, 95% CI: 1.05-1.26) and hemoglobin at ASCT (HR 0.80, 95% CI: 0.68-0.94) were independent factors influencing PFS. In the model for OS, the only statistically significant factors were NLR at ASCT (HR 1.15, 95% CI: 1.04-1.27), bortezomib administration prior to ASCT (HR 0.52, 95% CI: 0.33-0.83) and age at diagnosis (HR 1.03, 95% CI: 1.00-1.06). NLR at ASCT is an independent predictive factor in MM patients undergoing ASCT.
Collapse
Affiliation(s)
- Damian Mikulski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland.,Department of Hematology, Copernicus Memorial Hospital in Lodz Comprehensive Cancer Center and Traumatology, Lodz, Poland
| | - Kacper Kościelny
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Mateusz Nowicki
- Department of Hematology, Copernicus Memorial Hospital in Lodz Comprehensive Cancer Center and Traumatology, Lodz, Poland.,Department of Hematology, Medical University of Lodz, Lodz, Poland
| | - Ewa Wawrzyniak
- Department of Hematology, Medical University of Lodz, Lodz, Poland
| | - Marta Kalwas
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Lodz, Poland
| | - Monika Kowalik
- Department of Hematology, Copernicus Memorial Hospital in Lodz Comprehensive Cancer Center and Traumatology, Lodz, Poland
| | - Mateusz Pryt
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Emilia Sęczkowska
- Department of Anesthesiology and Intensive Medical Therapy, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Agnieszka Świątek
- Department of Internal Medicine and Gastroenterology, Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Agnieszka Wierzbowska
- Department of Hematology, Copernicus Memorial Hospital in Lodz Comprehensive Cancer Center and Traumatology, Lodz, Poland.,Department of Hematology, Medical University of Lodz, Lodz, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland.,Department of Radiation Oncology, Dana‑Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
266
|
Kerbauy MN, Arcuri LJ, Favareto SL, de Rezende ACP, Hamerschlak N. Total marrow irradiation in hematopoietic stem cell transplantation for hematologic malignancies. Front Med (Lausanne) 2023; 10:1155954. [PMID: 37153098 PMCID: PMC10157478 DOI: 10.3389/fmed.2023.1155954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/23/2023] [Indexed: 05/09/2023] Open
Abstract
Total body irradiation (TBI) has been an essential component of the conditioning regimen in hematopoietic cell transplantation for many years. However, higher doses of TBI reduce disease relapse at the expense of more significant toxicities. Therefore, total marrow irradiation and total marrow and lymphoid irradiation have been developed to deliver organ-sparing targeted radiotherapy. Data from different studies show that TMI and TMLI can be safely administered in escalating doses in association with different chemotherapy conditioning regimen protocols, in situations with unmet needs, such as multiple myeloma, high-risk hematologic malignancies, relapsed or refractory leukemias, and elderly or frail patients, with low rates of transplant-related mortality. We reviewed the literature on applying TMI and TMLI techniques in autologous and allogeneic hematopoietic stem cell transplantation in different clinical situations.
Collapse
Affiliation(s)
- Mariana Nassif Kerbauy
- Department of Hematology and Bone Marrow Transplantation, Hospital Israelita Albert Einstein, São Paulo, Brazil
- *Correspondence: Mariana Nassif Kerbauy
| | | | | | | | - Nelson Hamerschlak
- Department of Hematology and Bone Marrow Transplantation, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
267
|
Fuchs O. Targeting cereblon in hematologic malignancies. Blood Rev 2023; 57:100994. [PMID: 35933246 DOI: 10.1016/j.blre.2022.100994] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 01/28/2023]
Abstract
The protein cereblon (CRBN) is a substrate receptor of the cullin 4-really interesting new gene (RING) E3 ubiquitin ligase complex CRL4CRBN. Targeting CRBN mediates selective protein ubiquitination and subsequent degradation via the proteasome. This review describes novel thalidomide analogs, immunomodulatory drugs, also known as CRBN E3 ubiquitin ligase modulators or molecular glues (avadomide, iberdomide, CC-885, CC-90009, BTX-1188, CC-92480, CC-99282, CFT7455, and CC-91633), and CRBN-based proteolysis targeting chimeras (PROTACs) with increased efficacy and potent activity for application in hematologic malignancies. Both types of CRBN-binding drugs, molecular glues, and PROTACs stimulate the interaction between CRBN and its neosubstrates, recruiting target disease-promoting proteins and the E3 ubiquitin ligase CRL4CRBN. Proteins that are traditionally difficult to target (transcription factors and oncoproteins) can be polyubiquitinated and degraded in this way. The competition of CRBN neosubstrates with endogenous CRBN-interacting proteins and the pharmacology and rational combination therapies of and mechanisms of resistance to CRL4CRBN modulators or CRBN-based PROTACs are described.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 12800 Praha 2, Czech Republic.
| |
Collapse
|
268
|
Guerra MC, Jerez J, Godoy G, Briones JL, Torres C, Hidalgo S, Goldschmidt V, Gazitúa R. Survival and response deepening after autologous transplantation in patients with multiple myeloma in Chile. Hematology 2022; 27:1223-1229. [DOI: 10.1080/16078454.2022.2142401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Joaquín Jerez
- Department of Haematology, Fundación Arturo López Pérez, Providencia, Chile
- Resident of Haematology, Universidad de los Andes, Santiago, Chile
| | - Giselle Godoy
- Department of Haematology, Fundación Arturo López Pérez, Providencia, Chile
| | - José Luis Briones
- Department of Haematology, Fundación Arturo López Pérez, Providencia, Chile
| | - Carlos Torres
- Department of Haematology, Fundación Arturo López Pérez, Providencia, Chile
| | - Sebastián Hidalgo
- Department of Haematology, Fundación Arturo López Pérez, Providencia, Chile
| | | | - Raimundo Gazitúa
- Department of Haematology, Fundación Arturo López Pérez, Providencia, Chile
| |
Collapse
|
269
|
Geng C, Zhou H, Wang H, Li Y, Leng Y, Zhang Z, Jian Y, Yang G, Chen W. Newly diagnosed multiple myeloma patients with CD56 expression benefit more from autologous stem cell transplantation. BMC Cancer 2022; 22:1349. [PMID: 36564753 PMCID: PMC9783713 DOI: 10.1186/s12885-022-10382-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/29/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Several studies showed that lack of CD56 expression was a poor prognostic factor for patients with newly diagnosed multiple myeloma (NDMM). However, other studies were not able to confirm the prognostic value of CD56 in NDMM. This study aimed to evaluate the prognostic value of CD56 expression for patients with NDMM who received autologous stem cell transplantation (ASCT). METHODS We retrospectively analyzed 370 patients with NDMM under 66 years old and the propensity score matching technique was used to reduce the bias between two groups. RESULTS CD56 expression was observed in 250 (67.6%) patients, and only half of transplant-eligible patients received ASCT for financial and adverse effects concerns after induction therapy. 54.8% (137/250) CD56 positive patients received ASCT; and 47.5% (57/120) CD56 negative patients received ASCT. Univariate and multivariate analyses showed that ASCT was correlated with longer overall survival (OS) (p < 0.001) and progression-free survival (PFS) (p < 0.001) for CD56 positive patients. However, ASCT had no impact on OS and PFS in univariate and multivariate analysis (p > 0.05). In the propensity score matching analysis, 186 CD56 positive patients were identified, 93 patients had received ASCT and 93 patients had no ASCT. Among 120 CD56 negative patients, 80 patients, 40 in each group, were identified. Among 186 matched CD56 positive patients, patients with ASCT had longer OS (87.6 vs.56.1 months, p = 0.049) and PFS (36.7 vs.30.9 months, p = 0.040). However, ASCT had no impact on OS and PFS for matched CD56 negative patients (p > 0.05). CONCLUSIONS These results demonstrated that ASCT may improve OS and PFS of CD56 positive patients and had no impact on survival of CD56 negative patients.
Collapse
Affiliation(s)
- Chuanying Geng
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Huixing Zhou
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Huijuan Wang
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Yanchen Li
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Yun Leng
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Zhiyao Zhang
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Yuan Jian
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Guangzhong Yang
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| | - Wenming Chen
- grid.24696.3f0000 0004 0369 153XDepartment of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020 China
| |
Collapse
|
270
|
Offidani M, Morè S, Corvatta L. Maintenance therapy in multiple myeloma: Two is not always better than one. Br J Haematol 2022; 201:181-182. [PMID: 36541138 DOI: 10.1111/bjh.18614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
In multiple myeloma (MM) long-term therapy aims to control disease and delay progression for as long as possible. In this issue Jenner et al. failed to demonstrate a benefit of maintenance with lenalidomide plus vorinostat compared with lenalidomide in both transplant eligible (TE) and ineligible (NTE) patients enrolled in the Myeloma XI trial. Commentary on: Jenner et al. The addition of vorinostat to lenalidomide maintenance for patients with newly diagnosed multiple myeloma of all ages: results from 'Myeloma XI', a multicentre, open-label, randomised, phase III trial. Br J Haematol. 2023;201:276-288.
Collapse
Affiliation(s)
- Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona Ancona Italy
| | - Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona Ancona Italy
| | | |
Collapse
|
271
|
The first relapse in multiple myeloma: how to pick the next best thing. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:560-568. [PMID: 36485087 PMCID: PMC9821240 DOI: 10.1182/hematology.2022000356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The choice of treatment for patients with multiple myeloma (MM) at first relapse/progression is based on many factors: (1) treatment-related factors, which include the regimen used during first induction, the quality and duration of first response achieved, toxicities from the first treatment, whether the patient underwent autologous stem cell transplant, and whether the patient was on maintenance at the time of relapse/progression; (2) disease-related factors, including disease presentation and pace of progression; and (3) patient-related factors, including functional age and performance status. The learning objectives are to present the treatment options for patients with MM upon their first relapse and to learn about various strategies for selecting an optimal treatment regimen.
Collapse
|
272
|
Costello CL. Newly diagnosed multiple myeloma: making sense of the menu. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:539-550. [PMID: 36485145 PMCID: PMC9820388 DOI: 10.1182/hematology.2022000404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The development of new drugs and subsequent novel combinations for the treatment of newly diagnosed multiple myeloma (NDMM) has resulted in a plethora of treatment options that can make the choice of initial induction therapy a challenge. A greater understanding of both patient- and disease-specific factors can provide a personalized approach to help design a treatment course. Historically, the choice of an induction regimen has been tethered to an initial impression of transplant eligibility at the time of diagnosis. As more effective and better-tolerated induction regimens have emerged, there has been increasing overlap in the induction strategies used for all patients with NDMM, which increasingly provide the ultimate goal of deep and durable remissions. The current treatment options and strategies for the management of NDMM are evaluated using the best available data to provide a rationale for these decisions.
Collapse
Affiliation(s)
- Caitlin L. Costello
- Correspondence Caitlin L. Costello, UCSD, 3855 Health Sciences Drive, MC 0960, La Jolla, CA 92093, USA; e-mail:
| |
Collapse
|
273
|
Pawlyn C, Khan AM, Freeman CL. Fitness and frailty in myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:337-348. [PMID: 36485137 PMCID: PMC9820647 DOI: 10.1182/hematology.2022000346] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As the aging population grows, so too does the number of well-tolerated antimyeloma therapies. Physicians will see an increasing volume of patients for subsequent lines of therapy, which could now extend this relationship for over a decade. For younger patients, treatment choices are infrequently impacted by concerns of fitness, but instead about effecting the deepest, most durable response. Older adults, in contrast, are more likely to experience under- than overtreatment, and therefore more objective (and ideally straightforward) ways to evaluate their fitness and ability to tolerate therapy will increasingly assist in decision-making. Post hoc analyses categorizing the fitness of trial patients in the modern treatment era globally demonstrate that even in highly selected populations, those that are recategorized as less fit or frail are consistently at higher risk of inferior outcomes and increased toxicities. Real-world data are comparatively lacking but do demonstrate that most patients with myeloma are not representative of those enrolled on clinical trials, generally more heavily burdened by comorbidities and more likely to be categorized as "less than fit." Simultaneously, the number of therapeutic options open to patients in the relapsed setting continues to grow, now including T-cell engagers and cellular therapies, with their unique toxicity profiles. The aim of this review is to summarize the available data, highlight some of the approaches possible to easily assess fitness and how results might inform treatment selection, and illustrate ways that patients' condition can be optimized rather than lead to exclusion from the more complex therapies newly available.
Collapse
Affiliation(s)
- Charlotte Pawlyn
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Abdullah M Khan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Ciara L Freeman
- H. Lee Moffitt Cancer Centre & Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| |
Collapse
|
274
|
Nooka AK, Lonial S. EXABS-123-MM Additional Agents for RRMM - How Do/Will They Fit. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22 Suppl 2:S24-S26. [PMID: 36163819 DOI: 10.1016/s2152-2650(22)00649-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Ajay K Nooka
- Emory University Winship Cancer Institute, 1365B Clifton Rd NE, Atlanta, GA 30322, USA
| | - Sagar Lonial
- Emory University Winship Cancer Institute, 1365B Clifton Rd NE, Atlanta, GA 30322, USA
| |
Collapse
|
275
|
Mai EK. Vascular thrombotic events in the era of modern myeloma therapy. Br J Haematol 2022; 199:642-644. [PMID: 36120817 DOI: 10.1111/bjh.18452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
The treatment landscape in multiple myeloma (MM) has changed drastically in the past two decades with new treatment paradigms evolving. In this issue Sborov et al. provide the first study investigating the frequency, severity, onset of vascular thrombotic events (VTEs) and use of prophylactic therapies in a trial applying a contemporary myeloma treatment protocol and highlight that prevention of VTEs needs to be revisited in the era of modern myeloma treatment.
Collapse
Affiliation(s)
- Elias Karl Mai
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
276
|
Atrash S. Between two kingdoms (CAR-T and hematopoietic stem cell transplantation). Transplant Cell Ther 2022; 28:413-414. [DOI: 10.1016/j.jtct.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
277
|
Kazandjian D, Landgren O. Novel Quadruplets and the Age of Immunotherapies in the Treatment of Newly Diagnosed Multiple Myeloma. JAMA Oncol 2022; 8:1260-1262. [PMID: 35862054 DOI: 10.1001/jamaoncol.2022.2421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Dickran Kazandjian
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Ola Landgren
- Myeloma Division, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
278
|
Xie Y, Yang H, Yang C, He L, Zhang X, Peng L, Zhu H, Gao L. Role and Mechanisms of Tumor-Associated Macrophages in Hematological Malignancies. Front Oncol 2022; 12:933666. [PMID: 35875135 PMCID: PMC9301190 DOI: 10.3389/fonc.2022.933666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Mounting evidence has revealed that many nontumor cells in the tumor microenvironment, such as fibroblasts, endothelial cells, mesenchymal stem cells, and leukocytes, are strongly involved in tumor progression. In hematological malignancies, tumor-associated macrophages (TAMs) are considered to be an important component that promotes tumor growth and can be polarized into different phenotypes with protumor or antitumor roles. This Review emphasizes research related to the role and mechanisms of TAMs in hematological malignancies. TAMs lead to poor prognosis by influencing tumor progression at the molecular level, including nurturing cancer stem cells and laying the foundation for metastasis. Although detailed molecular mechanisms have not been clarified, TAMs may be a new therapeutic target in hematological disease treatment.
Collapse
|
279
|
Gogia A, Ganguly S. Triplet therapy, transplantation, and maintenance until progression in multiple myeloma. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_241_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|