1
|
An N, Li J, Luo P, Wang D, Zhang P, Shu C, Cai S, Yu Q, Wen X, Wang X, Mu W, Hu J, Li C. Key predictors of long-term outcomes in BCMA-targeted CAR-T therapy for relapsed/refractory multiple myeloma. J Transl Med 2025; 23:552. [PMID: 40380297 PMCID: PMC12084914 DOI: 10.1186/s12967-025-06543-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND B-cell maturation antigen (BCMA)-directed chimeric antigen receptor T-cell (CAR-T) therapy exhibits high response rates in patients with relapsed/refractory multiple myeloma (r/r MM). However, the specific factors that influence the response duration remain poorly understood. METHODS This single-centre, retrospective observational study included 56 patients with r/r MM who received BCMA CAR-T therapy (equecabtagene autoleucel) at Tongji Hospital, China. We analysed response rates and long-term clinical outcomes and identified key factors contributing to the long-term efficacy of BCMA CAR-T therapy. RESULTS At a median follow-up of 39.6 months, the overall response rate (ORR) was 96.4%. Among the patients, 96.4% (54 of 56) achieved minimal residual disease (MRD) negativity, whereas 80.4% (45 of 56) achieved complete response (CR) or stringent complete response (sCR). Poorer outcomes were observed in patients with triple exposure, high cytogenetic risk, or failure to achieve CR. Better outcomes were associated with a CAR-T cell persistence of at least six months and sustained MRD negativity. Prolonged MRD negativity was strongly correlated with longer progression-free survival (PFS), with median PFS durations of 58 months, 64 months, and not reached (NR) for patients who maintained MRD negativity for 12, 24, and 36 months, respectively. Patients who remained MRD-negative and progression-free exhibited higher CAR-T cell expansion peaks. Additionally, CAR-T cell persistence was positively correlated with the duration of MRD negativity duration, PFS, and overall survival (OS). CONCLUSIONS BCMA CAR-T therapy provides durable responses in a subset of patients with r/r MM. Early intervention may improve patient prognosis by promoting sustained MRD negativity, thus improving overall treatment outcomes. TRIAL REGISTRATION Trial registration Chinese Clinical Trial Registry, ChiCTR2000033946 ( https://www.chictr.org.cn/showproj.html?proj=53503 ), Registered June 18, 2020. Trial registration Chinese Clinical Trial Registry, ChiCTR1800018137 ( https://www.chictr.org.cn/showproj.html?proj=30653 ), Registered August 31, 2018.
Collapse
Affiliation(s)
- Ning An
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Juan Li
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Luo
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Peiling Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Chang Shu
- Office of Drug Clinical Trial, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Songbai Cai
- Nanjing IASO Biotherapeutics Ltd, Shanghai, China
| | - Qiuxia Yu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Xinyu Wen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Xinran Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Jianlin Hu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, P. R. China.
- Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
2
|
Zhou H, Chen W. The efficacy of new drug regimens in treating newly diagnosed high-risk cytogenetic multiple myeloma patients: a systematic literature review and meta-analysis. Front Med (Lausanne) 2025; 12:1575914. [PMID: 40432724 PMCID: PMC12106411 DOI: 10.3389/fmed.2025.1575914] [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: 02/13/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Multiple myeloma (MM) is a plasma cell malignancy comprising 10% of hematologic cancers, associated with bone marrow dysfunction and organ damage. High-risk cytogenetic MM patients, identified by specific genetic abnormalities, face poor outcomes despite recent advancements. Traditional treatments often prove inadequate, necessitating novel regimens. This review assesses the efficacy of emerging therapies-next-generation proteasome inhibitors, immunomodulatory drugs, and CD38-targeting agents-aimed at improving outcomes for this patient subset. Methods A systematic review and meta-analysis were performed, analyzing data from 18 randomized controlled trials (RCTs) involving high-risk MM patients treated with new drug combinations. Data extraction, quality assessment, and meta-analysis were conducted using a Bayesian fixed-effects model. Results For transplant-eligible patients, CD38-based therapies reduced progression or death risk by 33% during induction and 48% during maintenance. They improved progression-free survival (PFS) by 38% in induction and 57% in maintenance and increased minimal residual disease (MRD) negativity by 38%. Dual novel drug regimens also enhanced MRD negativity, but Elotuzumab and Ixazomib regimens showed limited impact. Carfilzomib-based therapies showed varying PFS and survival benefits. Conclusion CD38-targeted regimens notably improve outcomes in high-risk cytogenetic MM, especially for transplant-eligible patients, by reducing disease progression, enhancing PFS, and increasing MRD negativity. Dual novel regimens show promise in MRD improvements. These findings support the potential of tailored therapeutic strategies to optimize patient care.
Collapse
Affiliation(s)
| | - Wenming Chen
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Multiple Myeloma Research Center of Beijing, Beijing, China
| |
Collapse
|
3
|
Mao J, Xue L, Wang H, Zhou H, Zhu Y, Jia T, Cai Z, Zhao L, Zhao L, Wang Y, Wang J. Cytogenetic risk stratification combined with minimal residual disease status influences the therapeutic outcome and prognosis of multiple myelomas. Sci Rep 2025; 15:12545. [PMID: 40217083 PMCID: PMC11992232 DOI: 10.1038/s41598-025-97125-w] [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: 12/18/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
To explore the value of cytogenetic risk stratification combined with minimal residual disease (MRD) status in predicting the therapeutic efficacy and prognosis for multiple myeloma (MM). From January 2017 to December 2023, 73 cases of newly diagnosed MM were recruited. Cytogenetic risks were stratified according to the results of fluorescence in situ hybridization (FISH); MRD status and clinical data were analyzed. The progression-free survival (PFS) and overall survival (OS), and their influencing factors were evaluated. During the median follow-up period of 30 (4-65) months, the median progression-free survival (PFS) and overall survival (OS) were 38 (95% CI 29.7, 46.3) months and 55 (95% CI 45.9, 64.1) months, respectively. In our cohort, the 3-year PFS rate was 51.0% and the 3-year OS rate was 71.0%. According to the MRD status, 31 MM patients were assigned to the MRD-positivity group and 42 to the MRD-negativity group. Significant differences were detected in the median PFS (30 months vs. 45 months, χ2 = 7.747, P = 0.005) and OS (34 months vs. 59 months, χ2 = 8.683, P = 0.003) between groups. Subgroup analyses based on the cytogenetic risk stratification (standard risk [SR] and high risk [HR]) showed that MM patients in the SR/MRD-negativity subgroup did not reach the median PFS and OS, and the median PFS (42 months vs. 33 months, P = 0.093) and OS (59 months vs. 42 months, P = 0.703) were similar between the SR/MRD-positivity and HR/MRD-negativity subgroups. In comparison to the HR/MRD-negativity subgroup, the median PFS (20 months vs. 33 months, P = 0.031) and OS (33 months vs. 42 months, P = 0.032) were significantly shorter in the HR/MRD-positivity group. Multivariate analysis showed that MRD-positivity was an independent risk factor for PFS (HR 2.874, 95% CI 1.452, 5.689; P = 0.008) and OS (HR 3.504, 95% CI 1.599, 7.676; P = 0.002) of MM. MRD status is a powerful prognostic indicator of PFS and OS in MM, but its performance is inferior to cytogenetic risk stratification. More high-risk cytogenetic abnormalities (HRCAs) indicate a worse prognosis of MM, while MRD-negativity improves HRCA-associated prognosis of MM. We recommend a risk stratification by assessing MRD status combined with HRCAs in MM patients, thus favoring the design of individualized treatment.
Collapse
Affiliation(s)
- Jianping Mao
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Lianguo Xue
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Haiqing Wang
- Laboratory Department, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Hang Zhou
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Yuanxin Zhu
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Tao Jia
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Zhimei Cai
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Lina Zhao
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Lidong Zhao
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Ying Wang
- Department of Hematology, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China.
| | - Juan Wang
- Department of Pediatrics, The First People's Hospital of Lianyungang, The Affiliated Lianyungang Hospital of Xuzhou Medical University, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang Clinical College of Nanjing Medical University, No. 6 Zhenhua Road, Haizhou District, Lianyungang, 222000, Jiangsu, China.
| |
Collapse
|
4
|
Mettias S, ElSayed A, Moore J, Berenson JR. Multiple Myeloma: Improved Outcomes Resulting from a Rapidly Expanding Number of Therapeutic Options. Target Oncol 2025; 20:247-267. [PMID: 39878864 DOI: 10.1007/s11523-024-01122-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2024] [Indexed: 01/31/2025]
Abstract
Multiple myeloma (MM) is a bone-marrow-based cancer of plasma cells. Over the last 2 decades, marked treatment advances have led to improvements in the overall survival (OS) of patients with this disease. Key developments include the use of chemotherapy, immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies. MM remains incurable, with outcomes influenced by many factors, including age, sex, genetics, and treatment response. This review summarizes recent studies regarding monitoring and treatment of MM, emphasizing the efficacy of new therapies, the impact of maintenance treatments, and approaches for managing relapsed or refractory MM. The role of specific drug classes used to treat MM, including immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, and newer treatments such as chimeric antigen receptor T-cell therapies and bispecific antibodies are discussed. Combination therapies have significantly improved outcomes. Maintenance therapies, particularly with lenalidomide, have been effective in extending OS but lead to an increased risk of secondary cancers. Venetoclax, selinexor, and ruxolitinib have shown potential as new therapeutic options for patients with relapsed or refractory MM. Immune-based treatments, such as chimeric antigen receptor T-cell therapy and bispecific antibodies, mark a major advancement for heavily pretreated patients, although challenges remain related to cost, availability, and side effects. The treatment landscape for patients with MM has seen significant progress, with current therapies providing a longer OS and better quality of life. Future research should focus on optimizing these strategies, personalizing therapies, and exploring new therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - James R Berenson
- Berenson Cancer Center, West Hollywood, CA, USA.
- Institute for Myeloma and Bone Cancer Research, 9201 W. Sunset Boulevard, Suite 300, West Hollywood, CA, 90069, USA.
- ONCOtherapeutics, West Hollywood, CA, USA.
| |
Collapse
|
5
|
Li S, Liu J, Peyton M, Lazaro O, McCabe SD, Huang X, Liu Y, Shi Z, Zhang Z, Walker BA, Johnson TS. Multiple Myeloma Insights from Single-Cell Analysis: Clonal Evolution, the Microenvironment, Therapy Evasion, and Clinical Implications. Cancers (Basel) 2025; 17:653. [PMID: 40002248 PMCID: PMC11852428 DOI: 10.3390/cancers17040653] [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: 01/10/2025] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Multiple myeloma (MM) is a complex and heterogeneous hematologic malignancy characterized by clonal evolution, genetic instability, and interactions with a supportive tumor microenvironment. These factors contribute to treatment resistance, disease progression, and significant variability in clinical outcomes among patients. This review explores the mechanisms underlying MM progression, including the genetic and epigenetic changes that drive clonal evolution, the role of the bone marrow microenvironment in supporting tumor growth and immune evasion, and the impact of genomic instability. We highlight the critical insights gained from single-cell technologies, such as single-cell transcriptomics, genomics, and multiomics, which have enabled a detailed understanding of MM heterogeneity at the cellular level, facilitating the identification of rare cell populations and mechanisms of drug resistance. Despite the promise of advanced technologies, MM remains an incurable disease and challenges remain in their clinical application, including high costs, data complexity, and the need for standardized bioinformatics and ethical considerations. This review emphasizes the importance of continued research and collaboration to address these challenges, ultimately aiming to enhance personalized treatment strategies and improve patient outcomes in MM.
Collapse
Affiliation(s)
- Sihong Li
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Jiahui Liu
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Madeline Peyton
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Regenstrief Institute, Indianapolis, IN 46202, USA
| | - Olivia Lazaro
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
| | - Sean D. McCabe
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Xiaoqing Huang
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN 46202, USA
| | - Zanyu Shi
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Zhiqi Zhang
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Brian A. Walker
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN 46202, USA
| | - Travis S. Johnson
- Indiana Bioscience Research Institute, Indianapolis, IN 46202, USA
- School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Martello M, Solli V, Mazzocchetti G, Solimando AG, Bezzi D, Taurisano B, Kanapari A, Poletti A, Borsi E, Armuzzi S, Vigliotta I, Pistis I, Desantis V, Marzocchi G, Rizzello I, Pantani L, Mancuso K, Tacchetti P, Testoni N, Nanni C, Zamagni E, Cavo M, Terragna C. High level of circulating cell-free tumor DNA at diagnosis correlates with disease spreading and defines multiple myeloma patients with poor prognosis. Blood Cancer J 2024; 14:208. [PMID: 39609411 PMCID: PMC11605000 DOI: 10.1038/s41408-024-01185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/26/2024] [Accepted: 11/04/2024] [Indexed: 11/30/2024] Open
Abstract
Multiple myeloma (MM) is a plasma cell (PC) disorder characterized by skeletal involvement at the time of diagnosis. Recently, cell-free DNA (cfDNA) has been proven to recapitulate the heterogeneity of bone marrow (BM) disease. Our aim was to evaluate the prognostic role of cfDNA at diagnosis according to disease distribution, and to investigate the role of the MM microenvironment inflammatory state in supplying the release of cfDNA. A total of 162 newly diagnosed MM patients were screened using 18F-FDG PET/CT and assessed by ultra low-pass whole genome sequencing (ULP-WGS). High cfDNA tumor fraction (ctDNA) levels were correlated with different tumor mass markers, and patients with high ctDNA levels at diagnosis were more likely to present with metabolically active paraskeletal (PS) and extramedullary (EM) lesions. Moreover, we demonstrated that microenvironment cancer-associated fibroblast (CAFs)-mediated inflammation might correlate with high ctDNA levels. Indeed, a high cfDNA TF level at diagnosis predicted a poorer prognosis, independent of R-ISS III and 1q amplification; the inclusion of >12% ctDNA in the current R-ISS risk score enables a better identification of high-risk patients. ctDNA can be a reliable and less invasive marker for disease characterization, and can refine patient risk.
Collapse
Affiliation(s)
- Marina Martello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.
| | - Vincenza Solli
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Gaia Mazzocchetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Antonio Giovanni Solimando
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe‑J), Unit of Internal Medicine "Guido Baccelli", University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Davide Bezzi
- Nuclear Medicine Unit, AUSL Romagna, Ravenna, Italy
| | - Barbara Taurisano
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Ajsi Kanapari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Andrea Poletti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Enrica Borsi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Silvia Armuzzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Ilaria Vigliotta
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Ignazia Pistis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Vanessa Desantis
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe‑J), Section of Pharmacology, University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Giulia Marzocchi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Ilaria Rizzello
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Lucia Pantani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Katia Mancuso
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Paola Tacchetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Nicoletta Testoni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elena Zamagni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Michele Cavo
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Carolina Terragna
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.
| |
Collapse
|
7
|
Xu W, Liang X, Liu S, Yi X, Tian M, Yue T, Zhang Y, Yan Y, Lan M, Long M, Zhang N, Wang J, Sun X, Hu R, Zhu Y, Ma X, Cheng Y, Xu J, Dai Y, Jin F. Dynamics of minimal residual disease and its clinical implications in multiple myeloma: A retrospective real-life analysis. Clin Med (Lond) 2024; 24:100252. [PMID: 39362336 PMCID: PMC11525448 DOI: 10.1016/j.clinme.2024.100252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/30/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Minimal residual disease (MRD) testing is a promising approach to tailor the treatment of multiple myeloma (MM). However, several major concerns remain to be addressed before moving it into daily practice, most of which stem from the dynamic nature of the MRD status. Thus, it is crucial to understand the MRD dynamics and propose its clinical implications. METHODS We retrospectively analysed the data of patients with newly diagnosed MM (NDMM) who had flow cytometry-based MRD tests at multiple time points after initiation of therapy. The impact of undetectable MRD (including attainment, duration and loss) on clinical outcomes was analysed. RESULTS In a cohort of 220 patients with NDMM, attainment of MRD- offered favourable outcomes (P < 0.0001 for both progression-free survival (PFS) and overall survival (OS)), regardless of baseline risk factors. Notably, MRD- duration ≥12 months was associated with an 83 % (95 % confidence interval (CI), 0.09-0.34; P < 0.0001) or 69 % (95 % CI, 0.13-0.76; P = 0.0098) reduction in risk of progression/death or death, while the longer MRD- was sustained, the better the outcome was. Loss of MRD- led to poor PFS (hazard ratio (HR) 0.01, 95 % CI 0-0.06, P < 0.0001) and OS (HR 0.03, 95 % CI 0-0.24, P = 0.0008). Most patients (70 %) who lost MRD- status carried high-risk cytogenetic abnormalities (HRCAs). While MRD- was temporally inconsistent with conventional therapeutic responses (eg ≥ complete remission or very good partial response), it predicted disease progression or recurrence more robustly than the latter. Last, the predictive value of the MRD status was independent of baseline risk factors (eg high-risk cytogenetic abnormality, International Staging System (ISS) or Revised (R-)ISS staging). CONCLUSIONS Longitudinal assessment of MRD during the treatment course and follow-up is required for monitoring disease progression or relapse, to guide treatment decisions. Accordingly, a prospective study is currently ongoing to investigate the feasibility and benefit of the MRD-tailored therapy according to the longitudinal changes of the MRD status.
Collapse
Affiliation(s)
- Weiling Xu
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China; Radiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyue Liang
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Shanshan Liu
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xingcheng Yi
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengru Tian
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Tingting Yue
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yingjie Zhang
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yurong Yan
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Maozhuo Lan
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Mengtuan Long
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Nan Zhang
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jingxuan Wang
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoxiao Sun
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Rui Hu
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yufeng Zhu
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xintian Ma
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yue Cheng
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiayi Xu
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Fengyan Jin
- Haematology Department, First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
8
|
Pasvolsky O, Pasyar S, Bassett RL, Khan HN, Tanner MR, Bashir Q, Srour S, Saini N, Lin P, Ramdial J, Nieto Y, Lee HC, Patel KK, Kebriaei P, Thomas SK, Weber DM, Orlowski RZ, Shpall EJ, Champlin RE, Qazilbash MH. Impact of pretransplant minimal residual disease in patients with multiple myeloma and a very good partial response or better receiving autologous hematopoietic stem cell transplantation. Cancer 2024; 130:1663-1672. [PMID: 38127583 PMCID: PMC11009063 DOI: 10.1002/cncr.35171] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/20/2023] [Accepted: 10/13/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND The prognostic significance of minimal residual disease (MRD) status before autologous hematopoietic stem cell transplantation (autoHCT) in patients with multiple myeloma (MM) has not been clearly elucidated. METHODS Retrospective single-center study of adult MM patients who achieved ≥very good partial response (VGPR) after induction therapy from 2015 to 2021 received upfront autoHCT and had available pretransplant MRD status by next-generation flow cytometry. The cohort was divided into pretransplant MRD-negative (MRDneg) and MRD-positive (MRDpos) groups. RESULTS A total of 733 patients were included in our analysis; 425 were MRDneg and 308 MRDpos at autoHCT. In the MRDpos group, more patients had high-risk cytogenetic abnormalities (48% vs. 38%, respectively; p = .025), whereas fewer patients achieved ≥CR before autoHCT (14% vs. 40%; p < .001). At day 100 after autoHCT, 37% of the MRDpos versus 71% of the MRDneg achieved ≥CR, and at best posttransplant response 65% versus 88% achieved ≥CR, respectively. After a median follow-up of 27.6 months (range, 0.7-82.3), the median PFS was significantly shorter for patients in the MRDpos group compared to the MRDneg group: 48.2 months (95% confidence interval [CI], 0.3-80.5) versus 80.1 months (95% CI, 0.5-80.1), respectively (p < .001). There was no significant difference in overall survival between the two groups (p = .41). Pretransplant MRDpos status was predictive of shorter PFS in multivariate analysis (hazard ratio, 1.80; 95% CI, 1.31-2.46; p < .001). The impact of pretransplant MRD status was retained in most of the examined subgroups. CONCLUSIONS In patients achieving ≥VGPR to induction, pretransplant MRDpos status was associated with a lower CR rate after autoHCT and a shorter PFS.
Collapse
Affiliation(s)
- Oren Pasvolsky
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah Pasyar
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roland L. Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hina N. Khan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Hematology/Oncology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Mark R. Tanner
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qaiser Bashir
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samer Srour
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neeraj Saini
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yago Nieto
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hans C. Lee
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krina K. Patel
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sheeba K. Thomas
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Donna M. Weber
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert Z. Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E. Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Muzaffar H. Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
Ho M, Kourelis T. The burden of myeloma: novel approaches to disease assessment. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:356-362. [PMID: 36485143 PMCID: PMC9820131 DOI: 10.1182/hematology.2022000348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Novel therapies in multiple myeloma (MM) have increased the rates of conventional complete remission (CR) in patients. However, patients in CR can have highly heterogeneous outcomes. Novel and more sensitive methods of assessing residual disease burden after therapy will help prognosticate this group better and, ideally, allow individualized therapy adjustments based on response depth in the future. Here, we review novel bone marrow, peripheral blood, and imaging methods for assessing myeloma burden and discuss the opportunities and limitations of incorporating these in everyday clinical practice.
Collapse
Affiliation(s)
- Matthew Ho
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| | - Taxiarchis Kourelis
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, MN
| |
Collapse
|
10
|
Tam T, Smith E, Lozoya E, Heers H, Andrew Allred P. Roadmap for new practitioners to navigate the multiple myeloma landscape. Heliyon 2022; 8:e10586. [PMID: 36164513 PMCID: PMC9508512 DOI: 10.1016/j.heliyon.2022.e10586] [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: 06/27/2022] [Revised: 08/17/2022] [Accepted: 09/05/2022] [Indexed: 10/25/2022] Open
Abstract
Multiple myeloma (MM) is a blood cancer in which monoclonal plasma cells cause end organ damage resulting in hypercalcemia, renal failure, anemia, and bone lesions. MM is considered incurable, however, recent advances in treatment have improved survival. Historically, MM has been treated with immunomodulatory drugs (IMiDs), proteosome inhibitors (PIs), and corticosteroids. While newer therapeutic approaches such as monoclonal antibodies and cellular therapies have broadened the treatment horizon, the selection and sequencing of these therapies has become more complex. This review aims to help advanced practice providers navigate through the diagnosis, staging, treatment, and supportive care considerations in the MM space.
Collapse
Affiliation(s)
- Tiffany Tam
- Banner MD Anderson Cancer Center, 2946 E Banner Gateway Drive, Gilbert, AZ 85295, United States
| | - Eric Smith
- Banner MD Anderson Cancer Center, 2946 E Banner Gateway Drive, Gilbert, AZ 85295, United States
| | - Evelyn Lozoya
- Banner MD Anderson Cancer Center, 2946 E Banner Gateway Drive, Gilbert, AZ 85295, United States
| | - Hayley Heers
- Banner MD Anderson Cancer Center, 2946 E Banner Gateway Drive, Gilbert, AZ 85295, United States
| | - P Andrew Allred
- Banner MD Anderson Cancer Center, 2946 E Banner Gateway Drive, Gilbert, AZ 85295, United States
| |
Collapse
|
11
|
Suzuki T, Maruyama D, Iida S, Nagai H. Recent advances in the management of older adults with newly diagnosed multiple myeloma in Japan. Jpn J Clin Oncol 2022; 52:966-974. [PMID: 35830865 PMCID: PMC9486880 DOI: 10.1093/jjco/hyac111] [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: 03/22/2022] [Accepted: 06/25/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma is a cancer of plasma cells; the incidence rate of multiple myeloma is high among older adults. Although significant advances have been made in the clinical management of multiple myeloma driven by the introduction of novel drugs, such as proteasome inhibitors, immuno- modulators and antibodies, multiple myeloma remains incurable. Hence, the current therapeutic goal for multiple myeloma is to achieve long-term survival while maintaining a good quality of life. In this context, personalized treatment to balance the efficacy and safety of therapies is important, especially for older adults as they display diverse physical, cognitive or organ functioning. Furthermore, old age is also often associated with frailty. Several tools for evaluating frailty in older adults with multiple myeloma are now available, and frail patients defined by these tools have shown a poor prognosis and more treatment-related toxicities. In addition, it is important to evaluate other factors, such as the International Staging System, high-risk chromosomal abnormalities and treatment response, to predict the clinical course of patients. Further investigations are required to determine how these factors can optimize the treatment for multiple myeloma. In this review, we present a detailed account on the developments and issues related to the current treatment approaches for older adults with newly diagnosed multiple myeloma. We also discuss the ongoing phase III clinical study conducted by the lymphoma study group of the Japan Clinical Oncology Group, which targeted older adults with newly diagnosed multiple myeloma.
Collapse
Affiliation(s)
- Tomotaka Suzuki
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Dai Maruyama
- Department of Hematology Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirokazu Nagai
- Department of Hematology, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
12
|
Weisel K, Wadlund AO, Gungor G, Dergarabetian E, Pacheco C, Masurkar N, Rodriguez-Otero P. Real-world study on adoption of standard of care (SoC) for transplant-eligible newly diagnosed multiple myeloma (TE-NDMM) patients between 2017 and 2020/2021 across France, Germany, Spain, and Italy. Eur J Haematol 2022; 109:388-397. [PMID: 35775385 DOI: 10.1111/ejh.13821] [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: 03/21/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES This non-interventional observational study described the current standard-of-care (SoC) for transplant-eligible newly diagnosed multiple myeloma (TE-NDMM) patients in France, Germany, Spain, and Italy, and recorded the evolution in regimen adoption in distinct elements of frontline treatment during 2017-2020/2021. METHODS Clinical information on ongoing (I) or previous (II) TE-NDMM patients was extracted from the Cancerology database. Proportions of patients receiving regimens in each element and the evolution in regimen adoption were determined for the entire population and each country. RESULTS Most common induction regimens among I patients were VRd in France (75.3%) and Spain (44.1%), VTd in Italy (65.2%), and regimens other than VRd/VTd/VCd in Germany. Maintenance was ongoing/planned for 78.3%, 62.3%, 65.2%, and 61.4% patients in France, Germany, Spain, and Italy, respectively. Among II patients, VRd induction increased from 27.0% in 2017 to 65.7% in 2019 in France, remained relatively low in Spain and Germany, and not present in Italy. In Italy and Spain, VTd induction declined from 72.4% and 58.3% in 2017 to 52.8% and 17.3% in 2019, respectively. VCd induction in Germany declined from 85.2% in 2017 to 64.1% in 2019. CONCLUSION The use of bortezomib triplets in induction varied markedly over time and between selected countries.
Collapse
Affiliation(s)
- Katja Weisel
- University Medical Center Hamburg-Eppendorf, Dept. Oncology, Hematology, BMT with Section of Pneumology, Martinistrasse 52, Hamburg, Germany
| | | | - Guntug Gungor
- Janssen, Kavacık, Keçeli Plaza, Ertürk Sk. No:13, 34810 Beykoz/İstanbul, Turkey
| | - Eileen Dergarabetian
- Janssen-Cilag Limited, 50-100 Holmers Farm Way, High Wycombe, Buckinghamshire, United Kingdom
| | - Cécile Pacheco
- Cerner Enviza France SAS, 198 avenue de France, Paris, France
| | - Nihar Masurkar
- Cerner Enviza France SAS, 198 avenue de France, Paris, France
| | - Paula Rodriguez-Otero
- Department of Hematology, Clínica Universidad de Navarra, Av. de Pío XII, 36, Pamplona, Navarra, Spain
| |
Collapse
|
13
|
Cho H, Shin S, Chung H, Jang JE, Kim YR, Cheong JW, Min YH, Lee ST, Choi JR, Kim JS. Real-world data on prognostic value of measurable residual disease assessment by fragment analysis or next-generation sequencing in multiple myeloma. Br J Haematol 2022; 198:503-514. [PMID: 35505579 DOI: 10.1111/bjh.18211] [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: 02/09/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 01/12/2023]
Abstract
Measurable residual disease (MRD) negativity is a strong prognostic indicator in multiple myeloma (MM). However, the optimal use of MRD in daily clinical practice has been hampered by the limited feasibility of MRD testing. Therefore, we examined the clinical relevance of commercially available MRD modalities based on clonality assays by fragment analysis with IdentiClone® (n = 73 patients) and next-generation sequencing (NGS) with LymphoTrack® (n = 116 patients) in newly diagnosed patients with MM who received autologous stem cell transplantation (ASCT). MRD was assessed at the end of induction (pre-ASCT) and/or at 100 days after ASCT (post-ASCT). MRD could not predict survival when assessed by fragment analysis. However, NGS-based MRD negativity at pre- or post-ASCT was beneficial in terms of progression-free and overall survival. Moreover, NGS-based MRD negativity was independently associated with improved progression-free and overall survival, and MRD-positive patients both pre- and post-ASCT had worst outcome. Indeed, initial adverse prognostic features by high-risk cytogenetics could be mitigated upon achieving MRD negativity by NGS. We demonstrate the feasibility and clinical benefit of achieving MRD negativity by commercially available clonality-based MRD assays in MM and support incorporating NGS, but not fragment analysis, to tailor therapeutic strategies in real-world practice.
Collapse
Affiliation(s)
- Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Jang
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo Hong Min
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
Mack EKM, Hartmann S, Ross P, Wollmer E, Mann C, Neubauer A, Brendel C, Hoffmann J. Monitoring multiple myeloma in the peripheral blood based on cell-free DNA and circulating plasma cells. Ann Hematol 2022; 101:811-824. [PMID: 35106639 PMCID: PMC8913458 DOI: 10.1007/s00277-022-04771-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/16/2022] [Indexed: 11/25/2022]
Abstract
With the advent of novel, highly effective therapies for multiple myeloma (MM), classical serologic monitoring appears insufficient for response assessment and prediction of relapse. Moreover, serologic studies in MM are hampered by interference of therapeutic antibodies. The detection of malignant plasma cell clones by next generation sequencing (NGS) or multiparameter flow cytometry (MFC) circumvents these difficulties and can be performed in the peripheral blood (pB) by targeting circulating cell-free DNA (cfDNA) or circulating plasma cells (CPCs), thus also avoiding an invasive sampling procedure. Here, we applied NGS of VJ light chain (LC) rearrangements in cfDNA and MFC of magnetically-enriched CD138-positive CPCs (me-MFC) to investigate disease burden in unselected MM patients. Sequencing was successful for 114/130 (87.7%) cfDNA samples and me-MFC results were analyzable for 196/205 (95.6%) samples. MM clones were detectable in 38.9% of samples taken at initial diagnosis or relapse (ID/RD), but only in 11.8% of samples taken during complete remission (CR). Circulating MM plasma cells were present in 83.3% of ID/RD samples and 9.9% of CR samples. Residual disease assessment by NGS or me-MFC in samples taken during very good partial remission or CR was 80% concordant. Notably, 4/4 (NGS) and 5/8 (me-MFC) positive CR samples were from patients with oligo- or non-secretory myeloma. The time to progression was shorter if there was evidence of residual myeloma in the pB. Together, our findings indicate that our two novel analytical approaches accurately indicate the course of MM and may be particularly valuable for monitoring patients with serologically non-trackable disease.
Collapse
Affiliation(s)
- Elisabeth K M Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| | - Sören Hartmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Petra Ross
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Ellen Wollmer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Christoph Mann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Jörg Hoffmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| |
Collapse
|