1
|
Paiva B, Shi Q, Puig N, Cedena MT, Orfao A, Durie BGM, Munshi NC, San-Miguel J. Opportunities and challenges for MRD assessment in the clinical management of multiple myeloma. Nat Rev Clin Oncol 2025; 22:424-438. [PMID: 40195455 DOI: 10.1038/s41571-025-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/09/2025]
Abstract
Measurable residual disease (MRD) assessment is, from the methodological point of view, ready for prime time in multiple myeloma (MM). Abundant evidence underscores the value of MRD status determined using highly sensitive next-generation flow cytometry and next-generation sequencing tests in evaluating response to treatment and, therefore, prognosis in patients with this disease. MRD response assessment and monitoring might present a range of opportunities for individualized patient management. Moreover, the considerable amounts of high-quality and standardized MRD data generated in clinical trials have led to the acceptance of MRD negativity as an early end point for accelerated regulatory approval of treatments for MM. The data leave no doubt that the efficacy of new regimens in inducing deeper and durable MRD-negative responses is connected with prolonged survival. Yet, several evidential, technical and practical challenges continue to limit the implementation of MRD-guided treatment strategies in routine practice, and the use of MRD as a surrogate end point remains controversial to some. In this Review, we draw on past and present research to propose opportunities for overcoming some of these challenges, and to accelerate the use of MRD assessment for improved clinical management of patients with MM.
Collapse
Affiliation(s)
- Bruno Paiva
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain.
| | - Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Noemi Puig
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00233, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Maria-Teresa Cedena
- Instituto de Investigación imas12, CIBER-ONC number CB16/12/00369, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Alberto Orfao
- Department of Medicine and Cytometry Service, Centro de Investigación del Cancer (IBMCC, USAL-CSIC), CIBER-ONC number CB16/12/00400, University of Salamanca, Salamanca, Spain
| | - Brian G M Durie
- Division of Hematology/Oncology, Cedars-Sinai Outpatient Cancer Center, Los Angeles, CA, USA
| | - Nikhil C Munshi
- Basic and Correlative Science, Jerome Lipper Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, VA Boston Healthcare System, Boston, MA, USA
| | - Jesús San-Miguel
- Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Cancer Center Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Aljama MA, Sidiqi HM, Gertz MA. Are we maintaining minimal residual disease in myeloma? Leuk Lymphoma 2025; 66:1001-1009. [PMID: 39835888 DOI: 10.1080/10428194.2025.2455485] [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: 10/04/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Minimal residual disease (MRD) has emerged as an important prognostic maker in patients with multiple myeloma at different stages of their treatment. Moreover, it is being increasingly incorporated as an endpoint in various clinical trials. Since maintenance therapy is an integral part of myeloma treatment, especially in the upfront setting post autologous transplantation, it is imperative to understand the role of MRD testing in the maintenance stetting. This review aims to examine the utility and dynamics of MRD testing in order to elucidate its prognostic role and possible incorporation in clinical decision making processes.
Collapse
Affiliation(s)
| | - Hasib M Sidiqi
- Hematology Department, Fiona Stanley Hospital, Perth Western, Australia
- Curtin Medical School, Curtin University, Perth Western, Australia
| | - Morie A Gertz
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Terpos E, Malandrakis P, Ntanasis-Stathopoulos I, Kostopoulos IV, Eleutherakis-Papaiakovou E, Kanellias N, Spiliopoulou V, Migkou M, Fotiou D, Theodorakakou F, Kastritis E, Gavriatopoulou M, Tsitsilonis OE, Dimopoulos MA. Sustained bone marrow and imaging MRD negativity for 3 years drives discontinuation of maintenance post-ASCT in myeloma. Blood 2025; 145:2353-2360. [PMID: 40009496 DOI: 10.1182/blood.2024027686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/27/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRACT Discontinuation of lenalidomide maintenance after autologous stem cell transplantation is a burning question within the multiple myeloma (MM) community, especially after the inclusion of minimal residual disease (MRD) in the disease response criteria. In this prospective study, we evaluated the conversion to MRD positivity, the treatment-free survival (TFS), and the progression-free survival (PFS) in 52 patients with MM who discontinued lenalidomide maintenance after achieving sustained bone marrow and imaging MRD negativity for 3 years. Patients who developed MRD positivity after lenalidomide discontinuation restarted lenalidomide maintenance at the same dose. The median follow-up from lenalidomide discontinuation was 3 years. Overall, 12 (23%) patients obtained MRD positivity and restarted lenalidomide maintenance. Only 4 (7.6%) patients progressed; 3 had a biochemical progression and 1 had a clinical progression. The overall median PFS was not reached, whereas the 7-year PFS from diagnosis was 90.2%. The 1-, 2-, and 3-year TFS rates were 93.9%, 91.6%, and 75.8%, respectively, whereas the 1-, 2-, and 3-year landmark PFS rates from maintenance discontinuation (study entrance) were 96.0%, 96.0%, and 92.9%, respectively. There were no statistically significant associations among age, sex, Second Revision International Staging System, type of induction therapy, and use of consolidation therapy and the effect outcomes of PFS and TFS. We conclude that maintenance discontinuation after 3 years of sustained marrow and imaging MRD negativity is associated with low rates of MRD conversion and progressive disease. Thus, in the era of modern antimyeloma treatments, a subgroup of patients may remain treatment free while in complete remission without jeopardizing disease response.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis V Kostopoulos
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Nikolaos Kanellias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Spiliopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Magdalini Migkou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Fotiou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Foteini Theodorakakou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania E Tsitsilonis
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
4
|
Kündgen LJ, Akhoundova D, Hoffmann M, Legros M, Shaforostova I, Seipel K, Bacher U, Pabst T. Prognostic Value of Post-Transplant MRD Negativity in Standard Versus High- and Ultra-High-Risk Multiple Myeloma Patients. Cancers (Basel) 2025; 17:1565. [PMID: 40361491 PMCID: PMC12071325 DOI: 10.3390/cancers17091565] [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: 03/17/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Cytogenetic abnormalities and the persistence of minimal residual disease (MRD) following autologous stem cell transplantation (ASCT) are two established prognostically unfavorable biomarkers in multiple myeloma (MM). Previous studies have shown that post-transplant MRD status is a powerful predictor of progression-free survival (PFS) and overall survival (OS). However, the impact of MRD remains poorly characterized in MM patients with high- or ultra-high-risk cytogenetics. Objectives: This study investigated the prognostic value of post-transplant MRD in standard- versus high- and ultra-high-risk MM. To this aim, we performed a retrospective analysis of 137 MM patients who underwent high-dose chemotherapy (HDCT) and ASCT at our institution between January 2019 and December 2021. Cytogenetics were assessed by fluorescence in situ hybridization. High-risk genomic alterations included del(17p), t(4;14), t(14;16), t(14;20), gain(1q), and TP53 mutations, with two or more alterations defining the ultra-high-risk category. MRD was assessed in bone marrow aspirates post-ASCT using flow cytometry. Results: Eighty-two (60%) patients were categorized as being at standard risk, forty (29%) as high risk, and fifteen (11%) as ultra-high risk. Median follow-up was 47 months. MRD negativity was achieved in 76 (55%) patients. At 48 months, the overall PFS rate was 61% (72%, 50%, and 32% for the standard-, high-, and ultra-high-risk subgroups, respectively; p = 0.0004), while the OS rate was 85% (89%, 79%, and 80% in standard-, high-, and ultra-high-risk MM patients, respectively; p = 0.1494). Within the standard-risk subgroup, longer PFS was observed for patients achieving MRD negativity (p = 0.0172). High- and ultra-high-risk patients showed no significant differences in PFS when stratified by MRD status, possibly due to prompt progression to MRD positivity. Conclusions: Our results suggest that high- and ultra-high-risk MM patients might benefit from closer response monitoring, including dynamic MRD assessment. Further, high- and ultra-high-risk patients might require a more intensive peri-transplant treatment.
Collapse
Affiliation(s)
- Lea Jasmin Kündgen
- Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland; (L.J.K.); (D.A.); (M.H.); (I.S.); (K.S.)
| | - Dilara Akhoundova
- Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland; (L.J.K.); (D.A.); (M.H.); (I.S.); (K.S.)
| | - Michele Hoffmann
- Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland; (L.J.K.); (D.A.); (M.H.); (I.S.); (K.S.)
| | - Myriam Legros
- Department of Clinical Chemistry, Inselspital, University of Bern, CH-3010 Bern, Switzerland;
| | - Inna Shaforostova
- Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland; (L.J.K.); (D.A.); (M.H.); (I.S.); (K.S.)
| | - Katja Seipel
- Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland; (L.J.K.); (D.A.); (M.H.); (I.S.); (K.S.)
- Department for Biomedical Research (DBMR), University of Bern, CH-3008 Bern, Switzerland
| | - Ulrike Bacher
- Department of Hematology, Inselspital, University of Bern, CH-3010 Bern, Switzerland;
- Center of Laboratory Medicine (ZLM), Inselspital, University of Bern, CH-3010 Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, University of Bern, CH-3010 Bern, Switzerland; (L.J.K.); (D.A.); (M.H.); (I.S.); (K.S.)
| |
Collapse
|
5
|
Sachpekidis C, Goldschmidt H, Edenbrandt L, Dimitrakopoulou-Strauss A. Radiomics and Artificial Intelligence Landscape for [ 18F]FDG PET/CT in Multiple Myeloma. Semin Nucl Med 2025; 55:387-395. [PMID: 39674756 DOI: 10.1053/j.semnuclmed.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/16/2024]
Abstract
[18F]FDG PET/CT is a powerful imaging modality of high performance in multiple myeloma (MM) and is considered the appropriate method for assessing treatment response in this disease. On the other hand, due to the heterogeneous and sometimes complex patterns of bone marrow infiltration in MM, the interpretation of PET/CT can be particularly challenging, hampering interobserver reproducibility and limiting the diagnostic and prognostic ability of the modality. Although many approaches have been developed to address the issue of standardization, none can yet be considered a standard method for interpretation or objective quantification of PET/CT. Therefore, advanced diagnostic quantification approaches are needed to support and potentially guide the management of MM. In recent years, radiomics has emerged as an innovative method for high-throughput mining of image-derived features for clinical decision making, which may be particularly helpful in oncology. In addition, machine learning and deep learning, both subfields of artificial intelligence (AI) closely related to the radiomics process, have been increasingly applied to automated image analysis, offering new possibilities for a standardized evaluation of imaging modalities such as CT, PET/CT and MRI in oncology. In line with this, the initial but steadily growing literature on the application of radiomics and AI-based methods in the field of [18F]FDG PET/CT in MM has already yielded encouraging results, offering a potentially reliable tool towards optimization and standardization of interpretation in this disease. The main results of these studies are presented in this review.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Hartmut Goldschmidt
- Internal Medicine V, Hematology, Oncology and Rheumatology, German-Speaking Myeloma Multicenter Group (GMMG), Heidelberg University Hospital, Heidelberg, Germany
| | - Lars Edenbrandt
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
6
|
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
|
7
|
Mateos MV, Puig N. Accelerating Myeloma Drug Development: Will Minimal Residual Disease Replace Progression-Free Survival as an Intermediate Surrogate End Point? J Clin Oncol 2025; 43:1271-1274. [PMID: 40048684 DOI: 10.1200/jco-25-00048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/15/2025] [Accepted: 01/27/2025] [Indexed: 04/09/2025] Open
Affiliation(s)
- María-Victoria Mateos
- University Hospital of Salamanca
- Institute for Biomedical Research of Salamanca
- Institute of Cancer Molecular and Cellular Biology
- University of Salamanca, Salamanca, Spain
| | - Noemí Puig
- University Hospital of Salamanca
- Institute for Biomedical Research of Salamanca
- Institute of Cancer Molecular and Cellular Biology
| |
Collapse
|
8
|
Shi Q, Paiva B, Pederson LD, Dimier N, Talpes E, Prior TJ, Orfao A, Moreau P, Sonneveld P, Kumar SK, Dixon JG, Patel R, Bartlett BJ, Schecter J, McCarthy P, Hose D, Seckinger A, Mattia D, Goldschmidt H, Oliva S, Owen RG, Anderson KC, San-Miguel J, Durie BGM, Munshi N. Minimal Residual Disease-Based End Point for Accelerated Assessment of Clinical Trials in Multiple Myeloma: A Pooled Analysis of Individual Patient Data From Multiple Randomized Trials. J Clin Oncol 2025; 43:1289-1301. [PMID: 39938021 DOI: 10.1200/jco-24-02020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 02/14/2025] Open
Abstract
PURPOSE Newly approved drugs and combinations treating multiple myeloma (MM) have resulted in substantial improvements in patients' survival. To deliver rapid access to newer therapies, an earlier end point to expedite clinical trials is needed. Our objective was to evaluate the minimal residual disease-negative complete response (MRD-CR) as an intermediate end point for progression-free survival (PFS) and overall survival (OS) in newly diagnosed (ND) transplant-eligible (NDTE) patients, ND transplant-ineligible (NDTinE) patients, and patients with relapsed/refractory (RR) MM. PATIENTS AND METHODS Individual patient data from 20 randomized multicenter trials were collected. Eleven studies (4,773 patients) with sufficient data were analyzed to evaluate whether 9- or 12-month MRD-CR classified at a 10-5 threshold could be reasonably likely to predict the clinical benefit of new agents regarding PFS and OS. Global odds ratio (OR) was estimated using the bivariate Plackett Copula model. Supportive evaluation included correlations of the treatment effects on MRD-CR end points and PFS/OS, evaluated by both linear regression (R2weighted least squared) and Copula (R2Copula) models. RESULTS The analysis demonstrated that both 9- and 12-month MRD-CR strongly correlated with PFS at patient level in NDTE patients, NDTinE patients, and patients with RRMM. Global ORs ranged from 3.06 to 16.24, all with 95% CIs excluding 1.0. Encouraging trial-level correlations (R2, 0.61-0.70) were observed by pooling three populations and were stronger (R2, 0.67-0.78) in the ND population. Similar results were observed for OS. CONCLUSION Our findings provided the support for use of MRD-CR classified at a 10-5 threshold at either 9 or 12 months after starting of the treatment, as an intermediate end point to support accelerated approvals, in future trials in NDTE patients, NDTinE patients, and patients with RRMM.
Collapse
Affiliation(s)
- Qian Shi
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Bruno Paiva
- Department of Hematology and Immunology, University of Navara, Pamplona, Spain
| | - Levi D Pederson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | | | | | | | - Alberto Orfao
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Philippe Moreau
- Hematology Department, University Hospital Hotel-dieu, Nantes, France
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Jesse G Dixon
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Reshma Patel
- Johnson & Johnson Inc, High Wycombe, United Kingdom
| | | | | | | | - Dirk Hose
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), Jette, Belgium
| | - Anja Seckinger
- Department of Hematology and Immunology, Myeloma Center Brussels & Labor für Myelomforschung, Vrije Universiteit Brussel (VUB), Jette, Belgium
| | - D'Agostino Mattia
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, AOU Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Hartmut Goldschmidt
- University Hospital Heidelberg, GMMG-Study Group at the University Hospital Heidelberg, Heidelberg, Germany
| | - Stefania Oliva
- Myeloma Unit, Division of Hematology, University of Torino, Turin, Italy
| | | | | | - Jesús San-Miguel
- Professor of Hematology, Senior Consultant & Strategic Advisor, Clinica Universidad de Navarra, Pamplona, Spain
| | | | - Nikhil Munshi
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
9
|
Moukalled N, Malard F, Bazarbachi A, Mohty M. Minimal residual disease in multiple myeloma. Presse Med 2025; 54:104261. [PMID: 39662763 DOI: 10.1016/j.lpm.2024.104261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Minimal Residual Disease (MRD) in multiple myeloma has emerged as a significant prognostic factor, guiding treatment strategies and enhancing patient outcomes. Despite advancements in therapies such as proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, CAR-T cell therapy, and bispecific antibodies, complete eradication of malignant plasma cells remains challenging. MRD refers to a small number of residual cancer cells that persist after treatment and require sensitive methods like next-generation flow cytometry (NGF) and next-generation sequencing (NGS) for detection. MRD negativity has been associated with improved progression-free survival (PFS) and overall survival (OS), making it a key marker in clinical trials. The clinical utility of MRD lies in its ability to predict outcomes, with sustained MRD negativity linked to prolonged survival. Furthermore, it will likely help in tailoring treatment approaches, such as therapy escalation for high-risk patients or de-escalation for those achieving MRD negativity. Despite its prognostic value, challenges remain in standardizing MRD testing, ensuring its widespread availability, and addressing variability in results based on different detection methods. Future research aims to refine MRD-guided treatment and explore novel detection techniques, such as liquid biopsies, to improve patient monitoring in multiple myeloma.
Collapse
Affiliation(s)
- Nour Moukalled
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Florent Malard
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mohamad Mohty
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France.
| |
Collapse
|
10
|
Ntanasis‐Stathopoulos I, Filippatos C, Ntanasis‐Stathopoulos A, Malandrakis P, Kastritis E, Tsitsilonis OE, Dimopoulos MA, Terpos E, Gavriatopoulou M. Evaluating Minimal Residual Disease Negativity as a Surrogate Endpoint for Treatment Efficacy in Multiple Myeloma: A Meta-Analysis of Randomized Controlled Trials. Am J Hematol 2025; 100:427-438. [PMID: 39784302 PMCID: PMC11803549 DOI: 10.1002/ajh.27582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
This meta-analysis examined the association between minimal residual disease (MRD) negativity and survival outcomes in 15 304 patients with multiple myeloma (MM) enrolled in randomized controlled trials published until June 2, 2024. Overall, there was a significant, negative and strong association between MRD negativity odds ratios and survival hazard ratios (β_PFS = -0.20, p < 0.001, β_OS = -0.12, p = 0.023). These associations remained significant for newly diagnosed patients (β_PFS = -0.35, p < 0.001), and they were consistent but not significant for relapsed/refractory patients (β_PFS = -0.06, p = 0.635). Sustained MRD negativity at 1 year was strongly correlated with prolonged PFS (β_PFS = -0.30, p < 0.001). In conclusion, this comprehensive meta-analysis supports MRD as a surrogate for survival in MM.
Collapse
Affiliation(s)
| | - Charalampos Filippatos
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | | | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Ourania E. Tsitsilonis
- Department of Biology, School of SciencesNational and Kapodistrian University of AthensAthensGreece
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of MedicineNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
11
|
Liang X, Xu W, Zhou F, Huang W, Yi X, Zhang Y, Yan Y, Zhang N, Wang J, Sun X, Hu R, Zhu Y, Ma X, Sun Y, Lan M, Long M, Kumar SK, Dai Y, Jin F. Dissecting the high-risk property of 1q gain/amplification in patients with newly diagnosed multiple myeloma. Am J Cancer Res 2025; 15:501-516. [PMID: 40084363 PMCID: PMC11897644 DOI: 10.62347/fxvh4425] [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: 09/12/2024] [Accepted: 01/27/2025] [Indexed: 03/16/2025] Open
Abstract
1q gain/amplification (1q+) is the most common cytogenetic abnormality (CA), with a frequency of 30-50% in patients with newly diagnosed multiple myeloma (NDMM). Although accumulating evidence supports 1q+ as a "high-risk" CA (HRCA), several issues remain to be addressed to understand its true prognostic property. We retrospectively analyzed a cohort of 934 patients with NDMM from three centers in China, who had baseline data available for 1q+ [including 1q21 gain (3 copies) and amplification (> 3 copies)] detected by fluorescence in situ hybridization in isolated CD138+ cells, and who received first-line treatment with novel agents including proteasome inhibitors, immunomodulatory drugs, or both. Minimal residue disease (MRD) was assessed using next-generation flow cytometry. In this cohort, 1q+ patients accounted for 53% of all patients. 1q+ patients were characterized by larger tumor burden, more advanced diseases, adverse complications, and frequent concurrence of other CAs (particularly HRCAs) at diagnosis. Concurrence of HRCAs [del(17p), t(4;14), and t(14;16); known as double-hit MM], but not standard-risk CA, markedly worsened the outcome of 1q+ patients, compared to those with 1q+ only (progression-free survival/PFS: hazard ratio/HR 1.63, 95% confidence interval/CI 1.21-2.20, P = 0.0013; overall survival/OS: HR 1.96, 95% CI 1.40-2.74, P < 0.0001). 1q+ modulated the risk levels defined by the Revised International Staging System (R-ISS). Although the overall response rate was not significantly different between patients with or without 1q+, fewer 1q+ patients achieved complete response or better and minimal residue disease negativity (MRD-). MRD- attainment substantially prolonged PFS (HR 4.03, 95% CI 2.59-6.29, P < 0.0001) and OS (HR 3.72, 95% CI 2.24-6.19, P < 0.0001) of 1q+ patients. While 1q+ patients had relatively shorter MRD- duration, sustained MRD- significantly improved the PFS and OS of 1q+ patients. Together, 1q+ is an HRCA and a major component of double-hit MM, while the risk-adapted and MRD-tailored therapy may best help manage this high-risk population.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Weiling Xu
- Department of Radiology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Fan Zhou
- Department of Hematology and Oncology, Shanghai Jing’an District Zhabei Central HospitalShanghai, China
| | - Wenyang Huang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeTianjin, China
| | - Xingcheng Yi
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Yingjie Zhang
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Yurong Yan
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Nan Zhang
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Jingxuan Wang
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Xiaoxiao Sun
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Rui Hu
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Yufeng Zhu
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Xintian Ma
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Yue Sun
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Maozhou Lan
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Mengtuan Long
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Shaji K Kumar
- Division of Hematology, Mayo Clinic College of MedicineRochester, MN, USA
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin UniversityChangchun, Jilin, China
| | - Fengyan Jin
- Department of Hematology, The First Hospital of Jilin UniversityChangchun, Jilin, China
| |
Collapse
|
12
|
Jung SH, Koh Y, Kim MK, Kim JS, Moon JH, Min CK, Yoon DH, Yoon SS, Lee JJ, Hong CM, Kang KW, Kwon J, Kim KH, Kim DS, Kim SY, Kim SH, Kim YR, Do YR, Mun YC, Park SS, Park YH, Shin HJ, Eom HS, Yoon SE, Hwang SM, Lee WS, Lee MW, Yi JH, Lee JY, Lee JH, Lee HS, Lim SN, Lim J, Yhim HY, Chang YH, Jo JC, Cho J, Cho H, Choi YS, Cho HJ, Ahn A, Choi JH, Kim HJ, Kim K. Evidence-based Korean guidelines for the clinical management of multiple myeloma: addressing 12 key clinical questions. Blood Res 2025; 60:9. [PMID: 39903326 PMCID: PMC11794900 DOI: 10.1007/s44313-025-00055-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Multiple myeloma (MM), a hematological malignancy, is characterized by malignant plasma cell proliferation in the bone marrow. Recent treatment advances have significantly improved patient outcomes associated with MM. In this study, we aimed to develop comprehensive, evidence-based guidelines for the diagnosis, prognosis, and treatment of MM. We identified 12 key clinical questions essential for MM management, guiding the extensive literature review and meta-analysis of the study. Our guidelines provide evidence-based recommendations by integrating patient preferences with survey data. These recommendations include current and emerging diagnostic tools, therapeutic agents, and treatment strategies. By prioritizing a patient-centered approach and rigorous data analysis, these guidelines were developed to enhance MM management, both in Korea and globally.
Collapse
Affiliation(s)
- Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Youngil Koh
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Kyoung Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Joon Ho Moon
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chang-Ki Min
- Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dok Hyun Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Soo Yoon
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Republic of Korea
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ka-Won Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jihyun Kwon
- Department of Internal Medicine, Hematology and Oncology, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyoung Ha Kim
- Division of Hematology and Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Dae Sik Kim
- Division of Oncology & Hematology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Sung Yong Kim
- Hematology & Oncology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Sung-Hyun Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Young Rok Do
- Division of Hematology-Oncology, Department of Internal Medicine, Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Yeung-Chul Mun
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Sung-Soo Park
- Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young Hoon Park
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Ho Jin Shin
- Division of Hematology-Oncology, Department of Internal Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Hyeon-Seok Eom
- Department of Hematology-Oncology, Center for Hematologic Malignancy, National Cancer Center, Goyang, Republic of Korea
| | - Sang Eun Yoon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, Republic of Korea
| | - Sang Mee Hwang
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Won Sik Lee
- Department of Internal Medicine, Hematology, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Myung-Won Lee
- Division of Hematology and Oncology, Department of Internal Medicine, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jun Ho Yi
- Division of Hematology-Oncology, Department of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Ji Yun Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Ji Hyun Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Ho Sup Lee
- Department of Internal Medicine, Kosin University College of Medicine, Kosin University Gospel Hospital, Busan, Republic of Korea
| | - Sung-Nam Lim
- Department of Internal Medicine, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Republic of Korea
| | - Jihyang Lim
- Department of Laboratory Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ho-Young Yhim
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Yoon Hwan Chang
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae-Cheol Jo
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Jinhyun Cho
- Division of Hematology-Oncology, Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Republic of Korea
| | - Hyungwoo Cho
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon Seok Choi
- Division of Hematology-Oncology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Jeong Cho
- Department of Hematology/Oncology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ari Ahn
- Department of Laboratory Medicine, College of Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Han Choi
- Department of Endocrine and Metabolism Medicine, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Hyun Jung Kim
- Institute for Evidence-Based Medicine, Cochrane Korea College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kihyun Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-Gu, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Ikeda D, Aikawa S, Misono C, Oura M, Fujii F, Sakuma H, Toho M, Uehara A, Tabata R, Narita K, Takeuchi M, Watari T, Otsuka Y, Matsue K. Soluble B-cell maturation antigen levels for disease monitoring in oligosecretory and nonsecretory relapsed multiple myeloma. Blood 2025; 145:526-532. [PMID: 39441915 DOI: 10.1182/blood.2024026028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/20/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT Soluble B-cell maturation antigen (sBCMA) is elevated on multiple myeloma (MM) cells. We investigated whether sBCMA levels correlated with other myeloma tumor volume indicators and its utility in monitoring oligosecretory/nonsecretory (O-S/Non-S) MM. In 115 patients with newly diagnosed MM, sBCMA was compared with M-protein levels, bone marrow plasma cells (BMPCs), circulating tumor cells (CTCs), and total diffusion volume (tDV; estimated by whole-body diffusion-weighted magnetic resonance imaging) at diagnosis. sBCMA levels increased significantly with International Staging System stage, chromosome 1q21 gain/amplification, and CTC levels. sBCMA also correlated strongly with %BMPC (r = 0.65) and moderately with tDV (r = 0.55) and paraprotein levels (involved immunoglobulin in IgG and IgA subtypes, r = 0.44 and 0.4; involved free light-chain levels in light-chain-only MM, r = 0.61, all P < .05). Longitudinal changes in sBCMA were consistent with disease status in both 17 O-S/Non-S and other secretory MM cases. Furthermore, sBCMA levels increased as early as 6 months prerelapse in almost all O-S/Non-S relapsed patients. Thus, sBCMA correlates strongly with total tumor volume in MM, as assessed using different modalities. We suggest that sBCMA is useful, not only for monitoring responses in patients with O-S/Non-S MM but also for early relapse detection and prediction.
Collapse
Affiliation(s)
- Daisuke Ikeda
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Shuichi Aikawa
- Department of Laboratory Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Chiho Misono
- Department of Laboratory Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Mitsuaki Oura
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Fuminari Fujii
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Hajime Sakuma
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Masanori Toho
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Atsushi Uehara
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Rikako Tabata
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Kentaro Narita
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Masami Takeuchi
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Tomohisa Watari
- Department of Laboratory Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Yoshihito Otsuka
- Department of Laboratory Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Chiba, Japan
| |
Collapse
|
14
|
Moreno DF, Oriol A, de la Rubia J, Hernández MT, Iñigo MB, Palomera L, de Arriba F, González Y, Teruel AI, Pardo JL, López de la Guía A, Sampol A, Ríos-Tamayo R, Sureda A, Gutiérrez NC, Calasanz MJ, Ramos MLM, Mateos MV, San Miguel J, Lahuerta JJ, Bladé J, Rosiñol L. Is t(11;14) Always a Standard-Risk Cytogenetic Abnormality? Results From GEM05MENOS65 and GEM2012 PETHEMA/GEM Transplantation Trials. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00024-2. [PMID: 39955256 DOI: 10.1016/j.clml.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/23/2025] [Indexed: 02/17/2025]
Abstract
PURPOSE Recent studies describe inferior outcomes in newly diagnosed multiple myeloma (NDMM) patients with t(11;14) treated with novel agents. MATERIALS AND METHODS We analyzed 240 NDMM transplant eligible (TE) patients who received triplet induction regimen in the GEM05MENOS65 (bortezomib, thalidomide and dexamethasone - VTD) and GEM2012 (bortezomib, lenalidomide and dexamethasone - VRD) clinical trials. RESULTS t(11;14) and standard risk (SR) non-t(11;14) were prevalent in 51 (21%) and 189 (79%) patients, respectively. Patients with t(11;14) treated with VTD had a lower overall response rate (ORR) (84% vs. 97%, P = .044) and lower negative minimal residual disease (MRD) (7.7% vs 35.1%, P = .049) after induction, as compared to SR non-t(11;14), while there were no differences in ORR (87% vs. 89%) or negative MRD (13.2% vs. 24.4%, P = .2) for these 2 subgroups in patients treated with VRD. The presence of t(11;14) impacted negatively on PFS in patients with VTD (hazard ratio 2.70; P = .005), while no differences were observed in those treated with VRD. CONCLUSION TE NDMM patients harboring t(11;14) had an inferior outcome compared with SR patients when receiving induction therapy with VTD while no differences were observed when receiving a lenalidomide containing regimen.
Collapse
Affiliation(s)
- David F Moreno
- Hospital Clínic de Barcelona, Hematology Department, IDIBAPS, Barcelona, Spain
| | - Albert Oriol
- Hematology Department, Institut Català d'Oncologia and Josep Carreras Research Institute, Hospital Germans Trias I Pujol, Badalona, Spain
| | - Javier de la Rubia
- Hematology Department, Hospital La Fe, Universidad Católica de Valencia and Centro de Investigación, Biomédica en Red de Cáncer, Valencia, Spain
| | - Miguel T Hernández
- Hematology Department, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | | | - Luis Palomera
- Hematology Department, Hospital Clínico Univesitario Lozano Blesa, Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain
| | - Felipe de Arriba
- Hematology Department, Hospital Universitario Morales Meseguer, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - Yolanda González
- Hospital Universitario Dr Josep Trueta, Hematology Department, Girona, Spain
| | - Ana Isabel Teruel
- Hospital Clínico Universitario de Valencia, Hematology Department, Valencia, Spain
| | | | | | - Antonia Sampol
- Hospital Son Espases, Hematology Department, Palma de Mallorca, Spain
| | | | - Anna Sureda
- Hematology Department, Institut Català d'Oncologia - L'Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Norma C Gutiérrez
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL). Cancer Research Center-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Maria-Jose Calasanz
- Cancer Center Clínica Universidad de Navarra, Hematology Department, CIMA, IDISNA, Pamplona, Spain
| | | | - María Victoria Mateos
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL). Cancer Research Center-IBMCC (USAL-CSIC), Salamanca, Spain
| | - Jesús San Miguel
- Cancer Center Clínica Universidad de Navarra, Hematology Department, CIMA, IDISNA, Pamplona, Spain
| | - Juan José Lahuerta
- Hospital Universitario Doce de Octubre, Hematology Department, Madrid, Spain
| | - Joan Bladé
- Hospital Clínic de Barcelona, Hematology Department, IDIBAPS, Barcelona, Spain
| | - Laura Rosiñol
- Hospital Clínic de Barcelona, Hematology Department, IDIBAPS, Barcelona, Spain.
| |
Collapse
|
15
|
Roy J, Cohen S, Sauvageau G, Ahmad I, Fournier V, Terra R, Caudrelier P, Thiant S, Thauvette G, Bambace N, Delisle JS, Lachance S, Kiss T, Bernard L, Roy DC, Veilleux O, LeBlanc R. A Pilot Study of UM171-Expanded Cord Blood Grafts for Tandem Auto/Allogeneic Hematopoietic Cell Transplant in High and Ultra-High-Risk Myeloma Patients. Transplant Cell Ther 2025; 31:34.e1-34.e14. [PMID: 39419177 DOI: 10.1016/j.jtct.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/12/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Multiple myeloma (MM) remains associated with a poor outcome, particularly in patients with advanced disease and high-risk (HR) cytogenetics. To date, the only curative treatment is allogeneic (allo) hematopoietic cell transplantation (HCT), but high incidences of graft versus host disease (GVHD), nonrelapse mortality (NRM) and disease progression remain important obstacles. Cord blood (CB) transplantation has been associated with low rates of relapse and chronic (c) GVHD, but its use has declined because of high incidences of infections, severe acute GVHD and high NRM. In other hematologic malignancies, UM171-expanded CB transplants have led to improved outcomes, allowing for the selection of smaller, better HLA-matched units. We aimed to investigate the safety and feasibility of single UM171-expanded single CB unit transplantation in frontline tandem auto/allo HCT for HR/ultra-HR MM patients. Newly diagnosed MM patients ≤ 65 years with an ISS stage II/III and del(17p), t(4;14), t(14;16), t(14;20), del(1p) or +1q, R-ISS 3, ≥ 2 cytogenetic abnormalities, or plasma cell leukemia without a sibling donor and availability of a 5-7/8 matched CB graft with ≥ 0.5 x 105 CD34+/kg and ≥ 1.5 x 107 TNCs/kg were eligible to this phase I/II prospective study (ClinicalTrials.gov NCT03441958). After induction and autologous HCT, patients received a reduced intensity conditioning regimen and were infused with 7-day UM171-expanded CD34+ cells, along with the lymphocytes contained in the CD34-negative fraction. The primary endpoints were feasibility of UM171 expansion, safety, kinetics of engraftment, incidences and maximum grades of acute and cGVHD at 1 and 2 years, assessment of measurable residual disease (MRD) and quality of life (QoL). Between 05/2018 and 11/2021, 20 patients were enrolled. One patient had an unsuccessful CB expansion with UM171, leaving 19 patients with a median age of 56 years. Median CD34+ cell dose infused after expansion was 4.62 x 106/kg (range: 0.79 to 5.76). Median times to achieve absolute neutrophil counts of 0.1 and 0.5 x 109/L were D+6 and D+10.5; median time to reach ≥ 20 x 109/L platelets was D+36. Full donor chimerism was achieved in all cell lineages by D+120 in recipients of reduced intensity conditioning. Cumulative incidences of grade II-IV, grade III-IV acute GVHD and moderate/severe cGVHD at 12 months were 68.4% (95% CI: 46 to 90), 5.3% (95% CI: 0% to 16%), and 10.5% (95% CI: 0% to 25%), respectively. With a median follow-up of 2.9 years (range: 0.46 to 5.3), cumulative incidences of relapse, PFS, OS and NRM at 3 years were 36.8% (95% CI: 14 to 59), 47.4% (95% CI: 29 to 76), 68.4% (95% CI: 50 to 93) and 15.8% (95%CI: 0 to 33), respectively. Median time to complete immunosuppression discontinuation was D+238. No unexpected adverse events were observed. Only one of 7 patients alive at 2 years with negative MRD at transplant has relapsed. Non-relapsing patients had a QoL after transplant similar to the general population. UM171-expanded CB transplant in HR/ultra-HR myeloma patients is feasible and allows the use of single CB units with a low risk of cGVHD. Patients with negative pretransplant MRD might benefit most from a UM171-expanded CB transplant.
Collapse
Affiliation(s)
- Jean Roy
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.
| | - Sandra Cohen
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Guy Sauvageau
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Imran Ahmad
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Valentyn Fournier
- Université de Lille, CNRS, UMR 9193 - SCALab - Sciences Cognitives et Sciences Affectives, Lille, France
| | - Rafik Terra
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada
| | | | - Stéphanie Thiant
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada
| | | | - Nadia Bambace
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Sébastien Delisle
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Silvy Lachance
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Thomas Kiss
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Léa Bernard
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Denis Claude Roy
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Olivier Veilleux
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Richard LeBlanc
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Montréal, Québec, Canada; Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
16
|
Muronova L, Soucek O, Zihala D, Sevcikova T, Popkova T, Plonkova H, Venglar O, Pour L, Stork M, Rihova L, Bezdekova R, Minarik J, Látal V, Novak M, Jungova A, Dekojova T, Straub J, Spacek M, Rezacova V, Maisnar V, Radocha J, Hajek R, Jelinek T. Real-World Evidence on Prognostic Value of MRD in Multiple Myeloma Using Flow Cytometry. Eur J Haematol 2025; 114:155-163. [PMID: 39390851 PMCID: PMC11613619 DOI: 10.1111/ejh.14316] [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: 08/08/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
Minimal residual disease (MRD) is one of the most important prognostic factors in multiple myeloma (MM) and a valid surrogate for progression-free survival (PFS) and overall survival (OS). Recently, MRD negativity was approved as an early clinical endpoint for accelerated drug approval in MM. Nevertheless, there is limited evidence of MRD utility in real-world setting. In this retrospective multicenter study, we report outcomes of 331 newly diagnosed MM patients with MRD evaluation at Day+100 after autologous stem cell transplantation using flow cytometry with a median limit of detection of 0.001%. MRD negativity was reached in 47% of patients and was associated with significantly prolonged median PFS (49.2 months vs. 18.4 months; hazard ratios (HR) = 0.37; p < 0.001) and OS (not reached vs. 74.9 months; HR = 0.50; p = 0.007). Achieving MRD negativity was associated with PFS improvements regardless of age, International Staging System (ISS) stage, lactate dedydrogenase (LDH) level, or cytogenetic risk. Importantly, MRD positive patients benefited from lenalidomide maintenance versus no maintenance (18-months PFS: 81% vs. 46%; HR = 0.24; p = 0.002) while in MRD negative patients such benefit was not observed (p = 0.747). The outcomes of our real-world study recapitulate results from clinical trials including meta-analyses and support the idea that MRD positive patients profit more from lenalidomide maintenance than MRD negative ones.
Collapse
Affiliation(s)
- Ludmila Muronova
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
| | - Ondrej Soucek
- Institute of Clinical Immunology and Allergology, Faculty of MedicineUniversity Hospital and Charles UniversityHradec KraloveCzech Republic
| | - David Zihala
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
- Department of Biology and Ecology, Faculty of ScienceUniversity of OstravaOstravaCzech Republic
| | - Tereza Sevcikova
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
- Department of Biology and Ecology, Faculty of ScienceUniversity of OstravaOstravaCzech Republic
| | - Tereza Popkova
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
| | - Hana Plonkova
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
| | - Ondrej Venglar
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
| | - Ludek Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Martin Stork
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Lucie Rihova
- Department of Clinical HematologyUniversity Hospital BrnoBrnoCzech Republic
| | - Renata Bezdekova
- Department of Clinical HematologyUniversity Hospital BrnoBrnoCzech Republic
| | - Jiri Minarik
- Department of Hemato‐Oncology, University Hospital Olomouc and Faculty of Medicine and DentistryPalacky UniversityOlomoucCzech Republic
| | - Vojtech Látal
- Department of Hemato‐Oncology, University Hospital Olomouc and Faculty of Medicine and DentistryPalacky UniversityOlomoucCzech Republic
| | - Martin Novak
- Department of Hemato‐Oncology, University Hospital Olomouc and Faculty of Medicine and DentistryPalacky UniversityOlomoucCzech Republic
| | - Alexandra Jungova
- Hematology and Oncology DepartmentCharles University Hospital PilsenPilsenCzech Republic
| | - Tereza Dekojova
- Hematology and Oncology DepartmentCharles University Hospital PilsenPilsenCzech Republic
| | - Jan Straub
- 1st Medical Department—Clinical Department of Haematology of the First Faculty of Medicine and General Teaching Hospital Charles UniversityPragueCzech Republic
| | - Martin Spacek
- 1st Medical Department—Clinical Department of Haematology of the First Faculty of Medicine and General Teaching Hospital Charles UniversityPragueCzech Republic
| | - Vladimira Rezacova
- Institute of Clinical Immunology and Allergology, Faculty of MedicineUniversity Hospital and Charles UniversityHradec KraloveCzech Republic
| | - Vladimir Maisnar
- 4th Department of Internal Medicine—HematologyUniversity Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec KraloveHradec KraloveCzech Republic
| | - Jakub Radocha
- 4th Department of Internal Medicine—HematologyUniversity Hospital Hradec Kralove, Charles University, Faculty of Medicine in Hradec KraloveHradec KraloveCzech Republic
| | - Roman Hajek
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
| | - Tomas Jelinek
- Department of HematooncologyUniversity Hospital OstravaOstravaCzech Republic
- Department of Hematooncology, Faculty of MedicineUniversity of OstravaOstravaCzech Republic
| |
Collapse
|
17
|
Zabaleta A, Puig N, Cedena M, Oliver‐Caldes A, Perez JJ, Moreno C, Tamariz‐Amador L, Rodriguez‐Otero P, Prosper F, Gonzalez‐Calle V, López‐Corral L, Rey‐Búa B, Puertas B, Mirás F, Sánchez‐Pina JM, López‐Muñoz N, Juan M, González‐Navarro EA, Urbano Á, de Larrea CF, Blade J, Lahuerta J, Martinez‐Lopez J, Mateos M, San Miguel JF, Paiva B. Clinical significance of complete remission and measurable residual disease in relapsed/refractory multiple myeloma patients treated with T-cell redirecting immunotherapy. Am J Hematol 2025; 100:93-102. [PMID: 39548827 PMCID: PMC11625977 DOI: 10.1002/ajh.27526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/25/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024]
Abstract
The impact of measurable residual disease (MRD) in relapse/refractory multiple myeloma (RRMM) patients treated with T-cell redirecting immunotherapy is uncertain. We analyzed MRD dynamics using next-generation flow in 201 patients treated in clinical trials with chimeric antigen receptor (CAR) T cells and T-cell engagers (TCE). Achieving MRD negativity at 10-6 was associated with 89% reduction in the risk of progression and/or death. Survival outcomes were improved in patients with sustained versus transient MRD negativity and were dismal in those who remained MRD positive. The intent-to-treat MRD negative rates were higher in patients treated with CAR T cells versus TCE. However, among patients achieving MRD negativity, there were no differences in survival outcomes when stratified according to treatment with CAR T cells versus TCE. In multivariate analysis including the number of prior lines of treatment, International Staging System, cytogenetic risk, extramedullary disease and type of T-cell redirecting immunotherapy, only the complete remission (CR) and MRD statuses showed independent prognostic value for progression-free and overall survival. In conclusion, our study shows that deep and sustained MRD negative CR is the most relevant prognostic factor and should be considered as the treatment endpoint in RRMM patients treated with CAR T cells and TCE.
Collapse
Affiliation(s)
- Aintzane Zabaleta
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Noemi Puig
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | | | - Aina Oliver‐Caldes
- Hospital Clinic de Barcelona, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | - José J. Perez
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | - Cristina Moreno
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Luis‐Esteban Tamariz‐Amador
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Paula Rodriguez‐Otero
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Felipe Prosper
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Veronica Gonzalez‐Calle
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | - Lucía López‐Corral
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | - Beatriz Rey‐Búa
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | - Borja Puertas
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | - Fátima Mirás
- Servicio de Hematología y HemoterapiaHospital 12 de OctubreMadridSpain
| | | | | | - Manel Juan
- Hospital Clinic de Barcelona, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | | | - Álvaro Urbano
- Hospital Clinic de Barcelona, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | | | - Joan Blade
- Hospital Clinic de Barcelona, IDIBAPSUniversity of BarcelonaBarcelonaSpain
| | - Juan‐José Lahuerta
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | | | - Maria‐Victoria Mateos
- Hospital Universitario de SalamancaInstituto de Investigacion Biomedica de SalamancaSalamancaSpain
| | - Jesús F. San Miguel
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Bruno Paiva
- Cancer Center Clinica Universidad de Navarra (CCUN), Centro de Investigacion Medica Aplicada (CIMA)Instituto de Investigacion Sanitaria de Navarra (IdiSNA)PamplonaSpain
| |
Collapse
|
18
|
Óskarsson JÞ, Rögnvaldsson S, Thorsteinsdottir S, Long TE, Ólafsson A, Eythorsson E, Jónsson Á, Viðarsson B, Önundarson PT, Agnarsson BA, Pálmason R, Sigurðardóttir M, Þorsteinsdóttir I, Ólafsson Í, Harding SJ, Durie BGM, Love TJ, Kristinsson SY. The significance of free light-chain ratio in light-chain monoclonal gammopathy of undetermined significance: a flow cytometry sub-study of the iStopMM screening study. Blood Cancer J 2024; 14:221. [PMID: 39695077 DOI: 10.1038/s41408-024-01201-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Light-chain (LC) monoclonal gammopathy of undetermined significance (MGUS) is a precursor of multiple myeloma (MM) and related conditions. LC-MGUS is characterized by free light-chain (FLC) levels outside defined reference intervals, indirectly indicating underlying plasma cell (PC) monoclonality. Next-generation flow cytometry (NGF) was used to evaluate clonal PC presence in bone marrow (BM) samples from individuals with LC-MGUS in the iStopMM study, aiming to assess the predictive value of the FLC ratio for clonal PC presence and its prognostic implications. BM samples from 61 individuals with LC monoclonal gammopathy were analyzed. Clonal plasma cells were detected in 53.6% of LC-MGUS samples (n = 28) and in all samples from individuals with more advanced conditions (n = 33). The FLC ratio was predictive of clonal PC presence for kappa-involved FLC ratios (p < 0.05; n = 42), with an optimal cutoff of 3.15 (96.7% sensitivity, 91.7% specificity). Of 195 individuals with kappa-involved LC-MGUS in follow-up within the iStopMM study, none with FLC ratios >1.65 to 3.15 progressed to MM (n = 124), whereas 4/71 (5.6%) with FLC ratios >3.15 progressed over median follow-up of 55 months. These findings support using a kappa-involved FLC ratio cutoff of >3.15 to more accurately identify individuals at increased risk of developing symptomatic PC disorders.
Collapse
Affiliation(s)
- Jón Þórir Óskarsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland.
- Landspítali University Hospital, Reykjavík, Iceland.
| | - Sæmundur Rögnvaldsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| | - Sigrun Thorsteinsdottir
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Thorir Einarsson Long
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Skane University Hospital, Lund, Sweden
| | - Andri Ólafsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Elias Eythorsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| | | | | | | | | | - Róbert Pálmason
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
- Skane University Hospital, Lund, Sweden
| | | | | | | | | | - Brian G M Durie
- Samuel Oschin Comprehensive Cancer Institude, Cedars-Sinai Outpatient Cancer Center, Los Angeles, CA, USA
| | - Thorvardur Jon Love
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| | - Sigurdur Y Kristinsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| |
Collapse
|
19
|
Nanni C, Deroose CM, Balogova S, Lapa C, Withofs N, Subesinghe M, Jamet B, Zamagni E, Ippolito D, Delforge M, Kraeber-Bodéré F. EANM guidelines on the use of [ 18F]FDG PET/CT in diagnosis, staging, prognostication, therapy assessment, and restaging of plasma cell disorders. Eur J Nucl Med Mol Imaging 2024; 52:171-192. [PMID: 39207486 PMCID: PMC11599630 DOI: 10.1007/s00259-024-06858-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
We provide updated guidance and standards for the indication, acquisition, and interpretation of [18F]FDG PET/CT for plasma cell disorders. Procedures and characteristics are reported and different scenarios for the clinical use of [18F]FDG PET/CT are discussed. This document provides clinicians and technicians with the best available evidence to support the implementation of [18F]FDG PET/CT imaging in routine practice and future research.
Collapse
Affiliation(s)
- Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals (UZ) Leuven, 3000, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sona Balogova
- Nuclear Medicine, Comenius University, Bratislava, Slovakia
- Médecine Nucléaire, Hôpital Tenon, GH AP.SU, Paris, France
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nadia Withofs
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liege, Belgium
- GIGA-CRC in Vivo Imaging, University of Liege, Liege, Belgium
| | - Manil Subesinghe
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Bastien Jamet
- Médecine Nucléaire, CHU Nantes, F-44000, Nantes, France
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy.
| | - Davide Ippolito
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
- University of Milano-Bicocca, School of Medicine, Via Cadore 33, 20090, Monza, Italy
| | | | | |
Collapse
|
20
|
Cartagena J, Deshpande A, Rosenthal A, Tsang M, Hilal T, Rimsza L, Kurzrock R, Munoz J. Measurable Residual Disease in Mantle Cell Lymphoma: The Unbearable Lightness of Being Undetectable. Curr Oncol Rep 2024; 26:1664-1674. [PMID: 39641852 DOI: 10.1007/s11912-024-01620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE OF REVIEW This paper evaluates the benefits and limitations of detecting measurable residual disease (MRD) in mantle cell lymphoma (MCL) and assesses its prognostic value. It also aims to highlight the importance of detecting low MRD levels post-treatment and their application in clinical practice. RECENT FINDINGS Recent studies show that MRD levels predict relapse and survival outcomes in hematologic neoplasms, including MCL. RT-qPCR is currently the most used method due to its high reproducibility and sensitivity. Ideal MRD detection should be highly sensitive, cost-effective, and applicable to a wide demographic of patients. This paper concludes that MRD detection has prognostic value in MCL but faces limitations in sensitivity and specificity. Further research is needed to establish the significance of low MRD levels before integrating these methods into clinical practice. Improved MRD detection technologies and understanding their impact on clinical outcomes will guide better patient management in MCL.
Collapse
Affiliation(s)
- Julio Cartagena
- University of Puerto Rico School of Medicine, San Juan, PR, USA
| | | | - Allison Rosenthal
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Mazie Tsang
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Talal Hilal
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Lisa Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Razelle Kurzrock
- Michels Rare Cancers Research Laboratories, Froedtert and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Javier Munoz
- Department of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
21
|
De Ramón C, Rojas EA, Misiewicz‐Krzeminska I, Cardona‐Benavides IJ, Cuadrado M, Isidro I, Calasanz M, Fernandez M, García‐Sanz R, Puig N, Cedena MT, Paiva B, Rosiñol L, Martínez‐López J, Bladé J, Lahuerta JJ, San Miguel JF, Mateos MV, Corchete LA, Gutiérrez NC, GEM/PETHEMA cooperative study group. Expression profile of Bcl-2 family proteins in newly diagnosed multiple myeloma patients. Hemasphere 2024; 8:e70036. [PMID: 39678396 PMCID: PMC11645297 DOI: 10.1002/hem3.70036] [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: 04/24/2024] [Revised: 07/31/2024] [Accepted: 08/28/2024] [Indexed: 12/17/2024] Open
Abstract
Antiapoptotic Bcl-2 family proteins are involved in myeloma cell survival. To date, their expression in multiple myeloma (MM) patients has mostly been analyzed at the RNA level. In the present study, we quantified for the first time the protein expression of the Bcl2-family members using a capillary electrophoresis immunoassay in 120 newly diagnosed MM patients, aged ≤65 years, treated in the context of the PETHEMA/GEM2012 study. We found that the pattern of expression of Bcl-2 family proteins was highly heterogeneous among patients. Although cases with t(11;14) had significantly higher levels of Bcl-2/Bcl-xL and Bcl-2+Bim+Bax/Bcl-xL ratios than those without t(11;14), the presence of this translocation was not synonymous with such high levels of expression. Conversely, some patients with other genetic alterations also showed higher levels of those ratios. Survival analysis revealed that the high expression of Bad and Puma proteins was associated with significantly longer overall survival (p = 0.001 and p < 0.001, respectively). Bcl-2 protein ratios predicting sensitivity to venetoclax in vitro were also able to distinguish patients with shorter time to progression after triplet-based induction therapy and ASCT. This is the first study to assess the expression of the most important Bcl-2 family proteins by a quantitative method in a large set of MM patients according to their cytogenetic abnormalities. We shed light on the impact of these proteins on MM prognosis, which could help to consider the levels of proteins involved in apoptosis in the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Cristina De Ramón
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Elizabeta A. Rojas
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | | | - Ignacio J. Cardona‐Benavides
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Myriam Cuadrado
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Isabel Isidro
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - María‐José Calasanz
- Department of HematologyClínica Universidad de Navarra, Centro de Investigaciones Biomédicas Aplicadas (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - Manuela Fernandez
- Spanish National Cancer Research Center (CNIO)MadridSpain
- Department of Hematology, University Hospital 12 de Octubre; Medicine DepartmentComplutense University MadridMadridSpain
| | - Ramón García‐Sanz
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - Noemi Puig
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - M. Teresa Cedena
- Spanish National Cancer Research Center (CNIO)MadridSpain
- Department of Hematology, University Hospital 12 de Octubre; Medicine DepartmentComplutense University MadridMadridSpain
| | - Bruno Paiva
- Department of HematologyClínica Universidad de Navarra, Centro de Investigaciones Biomédicas Aplicadas (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA)PamplonaSpain
| | - Laura Rosiñol
- Department of HematologyHospital Clinic of Barcelona, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Joaquín Martínez‐López
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
- Spanish National Cancer Research Center (CNIO)MadridSpain
- Department of Hematology, University Hospital 12 de Octubre; Medicine DepartmentComplutense University MadridMadridSpain
| | - Joan Bladé
- Department of HematologyHospital Clinic of Barcelona, Instituto de Investigaciones Biomédicas August Pi I Sunyer (IDIBAPS)BarcelonaSpain
| | - Juan J. Lahuerta
- Department of HematologyUniversity Hospital 12 de OctubreMadridSpain
| | - Jesús F. San Miguel
- Department of HematologyClínica Universidad de Navarra, Centro de Investigaciones Biomédicas Aplicadas (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA)PamplonaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - María V. Mateos
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - Luis A. Corchete
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | - Norma C. Gutiérrez
- Department of Hematology, University Hospital of SalamancaInstitute of Biomedical Research of Salamanca (IBSAL)SalamancaSpain
- Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)
| | | |
Collapse
|
22
|
Shen Q, Gong X, Feng Y, Hu Y, Wang T, Yan W, Zhang W, Qi S, Gale RP, Chen J. Measurable residual disease (MRD)-testing in haematological cancers: A giant leap forward or sideways? Blood Rev 2024; 68:101226. [PMID: 39164126 DOI: 10.1016/j.blre.2024.101226] [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: 06/20/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
Measurable residual disease (MRD)-testing is used in many haematological cancers to estimate relapse risk and to direct therapy. Sometimes MRD-test results are used for regulatory approval. However, some people including regulators wrongfully believe results of MRD-testing are highly accurate and of proven efficacy in directing therapy. We review MRD-testing technologies and evaluate the accuracy of MRD-testing for predicting relapse and the strength of evidence supporting efficacy of MRD-guided therapy. We show that at the individual level MRD-test results are often an inaccurate relapse predictor. Also, no convincing data indicate that increasing therapy-intensity based on a positive MRD-test reduces relapse risk or improves survival. We caution against adjusting therapy-intensity based solely on results of MRD-testing.
Collapse
Affiliation(s)
- Qiujin Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Xiaowen Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yahui Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Saibing Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK.
| | - Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| |
Collapse
|
23
|
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
|
24
|
Song Y, Zhao R, Fu W, Zhao J, Wang Q, Zhang R. Evaluation of the Prognostic Value of the Mayo Additive Staging System and Minimal Residual Disease in Newly Diagnosed Multiple Myeloma Patients. Cancer Med 2024; 13:e70382. [PMID: 39494582 PMCID: PMC11532867 DOI: 10.1002/cam4.70382] [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: 06/27/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024] Open
Abstract
INTRODUCTION This study aimed to evaluate the prognostic value of the Mayo additive staging system (MASS) and minimal residual disease (MRD) in newly diagnosed multiple myeloma (NDMM) patients. METHODS A total of 238 NDMM patients were enrolled in Beijing Chaoyang Hospital. Fluorescence in-situ hybridization and next-generation flow cytometry were used to examine cytogenetic abnormalities and MRD, respectively. The patients were classified into three groups to compare the effects on progression-free survival (PFS). Univariate and multivariate analyses were applied to identify the survival-related factors. RESULTS For MASS group, the PFS was significant difference in MASS I, II, and III patients (p = 0.0006); the patients with sustained MRD-negative, non-sustained MRD-negative, sustained MRD-positive, and non-sustained MRD-positive were divided into Groups 1, 2, 3, and 4, respectively. The Group 1 patients had superior PFS than Groups 2 and 3 patients (p < 0.05), but no difference in PFS was observed for Group 2 versus Group 3, Group 2 versus Group 4, and Group 3 versus Group 4 patients. For the MASS and MRD groups, among Groups 2, 3, and 4, MASS I patients had a superior PFS, while III patients showed the opposite result. Strikingly, no difference in PFS for Group 1 patients regardless of the MASS stage was observed. Despite being in MASS II and III, the PFS of Group 1 patients was longer than those with the other three groups. Response to treatment was an independent prognostic factor for MM patients. CONCLUSION Patients with an accumulation of high-risk factors and MRD-positive have poor outcomes. Sustained MRD-negative may improve high-risk patients' prognoses.
Collapse
Affiliation(s)
- Yichuan Song
- Department of Clinical Laboratory, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Rui Zhao
- Department of Clinical Laboratory, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Wenxuan Fu
- Department of Clinical Laboratory, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Jing Zhao
- Department of Clinical Laboratory, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Qingtao Wang
- Department of Clinical Laboratory, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| | - Rui Zhang
- Department of Clinical Laboratory, Beijing Chao‐Yang HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
25
|
Lakhwani S, Mateos MV, Martínez-López J, Paiva B, Rosiñol Dachs L, Martínez R, Oriol A, Bargay J, González-Montes Y, Gironella M, Encinas C, Martín J, Jarque I, Granell M, Abella E, García-Mateo A, Hernández-Rivas JÁ, Ramila E, Krsnik I, Casado Montero LF, De Arriba F, Palomera L, Sampol A, Moraleda JM, Casanova M, Delgado P, Lafuente A, Amutio E, López-Martínez A, Altés A, Ruíz MÁ, Alegre A, Lopez-Anglada L, De La Cruz J, Alonso Fernández R, Bladé Creixenti J, Lahuerta JJ, San-Miguel J, Hernández MT. Immunoparesis recovery in newly diagnosed transplant ineligible multiple myeloma patients, an independent prognostic factor that complements minimal residual disease. Ann Hematol 2024:10.1007/s00277-024-06031-0. [PMID: 39438321 DOI: 10.1007/s00277-024-06031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Information on the prognostic value of immunoparesis (IP) recovery in multiple myeloma (MM) patients has been only generated in some observational and retrospective studies. We have evaluated the prognostic impact of IP recovery and its association with minimal residual disease (MRD) in a series of 113 newly diagnosed transplant-ineligible (NDTI) patients, that received fix duration treatment (18 cycles of VMP/lenalidomide-dexamethasone) within the PETHEMA/GEM2010MAS65 trial and who achieved CR or VGPR. Immunoglobulin levels were measured at diagnosis, at the end of treatment (after cycle 18th) and during subsequent follow up whereas MRD was analyzed only at the end of the treatment (after cycle 18th). We found that patients who had IP at diagnosis and recovered it during or after treatment had longer progression free survival (PFS) [p < 0.001; HR 0.32 (0.19-0.52)] and longer overall survival (OS) [p = 0.007; HR 0.40 (0.20-0.80)] compared to those who failed to recover it. When we analyzed IP recovery in MRD negative patients, we found that those cases with IP recovery had longer PFS [p = 0.007; HR 0.31 (0.13-0.76)] and longer OS [p = 0.012; HR 0.21 (0.06-0.80)] as compared to MRD negative patients but without IP recovery. In conclusion, IP recovery confers better prognosis in NDTI-MM patients with fixed duration treatment who achieve CR or VGPR and the prognostic value of MRD can be complemented when combined with IP recovery.
Collapse
Affiliation(s)
- Sunil Lakhwani
- Hospital Universitario de Canarias, Universidad de La Laguna, Tenerife, Spain.
| | | | - Joaquín Martínez-López
- Hospital Universitario 12 de Octubre, Universidad Complutense, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | | | | | - Albert Oriol
- Institut Català d'Oncologia, Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona, Spain
| | - Joan Bargay
- Hospital Son Llàtzer, IdIsBa, Palma de Mallorca, Spain
| | | | | | - Cristina Encinas
- Hospital General Universitario Gregorio Marañón, IiSGM, Madrid, Spain
| | - Jesús Martín
- Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Isidro Jarque
- Hospital Universitari i Politecnic La Fe, Valencia, Spain
| | | | | | | | | | | | - Isabel Krsnik
- Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Felipe De Arriba
- Hospital Morales Meseguer, IMIB-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - Luis Palomera
- Hospital Clínico Universitario "Lozano Blesa", Zaragoza, Spain
| | - Antonia Sampol
- Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - José María Moraleda
- Hospital Clínico Universitario Virgen de la Arrixaca, IMIB, Universidad de Murcia, Murcia, Spain
| | | | | | | | | | | | - Albert Altés
- Hospital Althaia, Xarxa Assistencial de Manresa, Manresa, Spain
| | | | | | - Lucia Lopez-Anglada
- Unidad de Terapias Avanzadas de la Consejería de Sanidad de la Comunidad de Madrid, Madrid, Spain
| | - Javier De La Cruz
- Instituto De Investigación Sanitaria, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Rafael Alonso Fernández
- Hospital Universitario 12 de Octubre, Universidad Complutense, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Juan-José Lahuerta
- Instituto De Investigación Sanitaria, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jesús San-Miguel
- Cancer Center Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona, Spain
| | | |
Collapse
|
26
|
Schmidt T, Gahvari Z, Callander NS. SOHO State of the Art Updates and Next Questions: Diagnosis and Management of Monoclonal Gammopathy of Undetermined Significance and Smoldering Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:653-664. [PMID: 38641486 DOI: 10.1016/j.clml.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 04/21/2024]
Abstract
Monoclonal proteins are common, with a prevalence in the United States around 5% and the incidence increases with age. Although most patients are asymptomatic, the vast majority of cases are caused by a clonal plasma cell disorder. Monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) are asymptomatic precursor conditions with variable risk of progression to multiple myeloma (MM). In recent years, significant progress has been made to better understand the factors that lead to the development of symptoms and progression to myeloma. In this review, we summarize the current diagnosis treatment guidelines for MGUS and SMM and highlight recent advances that underscore a shifting paradigm in the evaluation and management of plasma cell precursor conditions.
Collapse
Affiliation(s)
- Timothy Schmidt
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison WI
| | - Zhubin Gahvari
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison WI
| | - Natalie S Callander
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison WI.
| |
Collapse
|
27
|
Kostopoulos IV, Ntanasis-Stathopoulos I, Rousakis P, Malandrakis P, Panteli C, Eleutherakis-Papaiakovou E, Angelis N, Spiliopoulou V, Syrigou RE, Bakouros P, Dimitrakopoulou G, Fotiou D, Migkou M, Kanellias N, Paschalidis N, Gavriatopoulou M, Kastritis E, Dimopoulos MA, Tsitsilonis OE, Terpos E. Low circulating tumor cell levels correlate with favorable outcomes and distinct biological features in multiple myeloma. Am J Hematol 2024; 99:1887-1896. [PMID: 38860642 DOI: 10.1002/ajh.27414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/12/2024]
Abstract
There is growing interest in multiple myeloma (MM) circulating tumor cells (CTCs), but their rareness in peripheral blood (PB) and inconsistency in cutoffs question their clinical utility. Herein, we applied next-generation flow cytometry in 550 bone marrow (BM) and matched PB samples to define an optimal CTC cutoff for both transplant-eligible and transplant-ineligible newly diagnosed MM (NDMM) patients. Deep phenotyping was performed to investigate unique microenvironmental features associated with CTC dissemination. CTCs were detected in 90% of patients (median 0.01%; range: 0.0002%-12.6%) and increased levels associated with adverse features. Correlations were observed between high CTC percentages and a diffused MRI pattern, a distinct BM composition characterized by altered B-cell differentiation together with an expansion of effector cells and tumor-associated macrophages, as well as a greater phenotypic dissimilarity between BM and PB clonal cells. Progression-free survival (PFS) and overall survival (OS) gradually worsened with each logarithmic increment of CTCs. Conversely, NDMM patients without CTCs showed unprecedented outcomes, with 5-year PFS and OS rates of 83% and 97%, respectively. A cutoff of 0.02% CTCs was independent of the ISS, LDH, and cytogenetics in a multivariate analysis of risk factors for PFS. The 0.02% CTC cutoff synergized with the MGUS-like phenotype and the R-ISS for improving the risk stratification systems. MRD negativity was less frequent if CTCs were ≥0.02% at diagnosis, but whenever achieved, the poor prognosis of these patients was abrogated. This study shows the clinical utility of CTC assessment in MM and provides evidence toward a consensus cutoff for risk stratification.
Collapse
Affiliation(s)
- Ioannis V Kostopoulos
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Pantelis Rousakis
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Chrysanthi Panteli
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Nikolaos Angelis
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Spiliopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Rodanthi-Eleni Syrigou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Bakouros
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Dimitrakopoulou
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Fotiou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Magdalini Migkou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kanellias
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Paschalidis
- Mass Cytometry-CyTOF Laboratory, Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Ourania E Tsitsilonis
- Department of Biology, School of Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
28
|
Cui J, Li X, Deng S, Du C, Fan H, Yan W, Xu J, Li X, Yu T, Zhang S, Lv R, Sui W, Hao M, Du X, Xu Y, Yi S, Zou D, Cheng T, Qiu L, Gao X, An G. Identification of Therapy-Induced Clonal Evolution and Resistance Pathways in Minimal Residual Clones in Multiple Myeloma through Single-Cell Sequencing. Clin Cancer Res 2024; 30:3919-3936. [PMID: 38900040 PMCID: PMC11369626 DOI: 10.1158/1078-0432.ccr-24-0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/16/2024] [Accepted: 06/17/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE In multiple myeloma (MM), therapy-induced clonal evolution is associated with treatment resistance and is one of the most important hindrances toward a cure for MM. To further understand the molecular mechanisms controlling the clonal evolution of MM, we applied single-cell RNA sequencing (scRNA-seq) to paired diagnostic and posttreatment bone marrow (BM) samples. EXPERIMENTAL DESIGN scRNA-seq was performed on 38 BM samples from patients with monoclonal gammopathy of undetermined significance (n = 1), MM patients at diagnosis (n = 19), MM posttreatment (n = 17), and one healthy donor (HD). The single-cell transcriptome data of malignant plasma cells (PC) and the surrounding immune microenvironment were analyzed. RESULTS Profiling by scRNA-seq data revealed three primary trajectories of transcriptional evolution after treatment: clonal elimination in patients with undetectable minimal residual disease (MRD-) and clonal stabilization and clonal selection in detectable MRD (MRD+) patients. We noted a metabolic shift toward fatty acid oxidation in cycling-resistant PCs, whereas selective PCs favored the NF-κB pathway. Intriguingly, when comparing the genetic and transcriptional dynamics, we found a significant correlation between genetic and nongenetic factors in driving the clonal evolution. Furthermore, we identified variations in cellular interactions between malignant PCs and the tumor microenvironment. Selective PCs showed the most robust cellular interactions with the tumor microenvironment. CONCLUSIONS These data suggest that MM cells could rapidly adapt to induction treatment through transcriptional adaptation, metabolic adaptation, and specialized immune evasion. Targeting therapy-induced resistance mechanisms may help to avert refractory disease in MM.
Collapse
Affiliation(s)
- Jian Cui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Xiaoyun Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Huishou Fan
- Department of Hematology, Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Wenqiang Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Jingyu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Xiaoqing Li
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Tengteng Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Shuaishuai Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Rui Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Xin Du
- Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
- Institute of Multiple Myeloma, Beijing GoBroad Boren Hospital, Beijing, China.
| | - Xin Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
- Institute of Multiple Myeloma, Beijing GoBroad Boren Hospital, Beijing, China.
| |
Collapse
|
29
|
Kubicki T, Dytfeld D, Barnidge D, Sakrikar D, Przybyłowicz-Chalecka A, Jamroziak K, Robak P, Czyż J, Tyczyńska A, Druzd-Sitek A, Giannopoulos K, Wróbel T, Nowicki A, Szczepaniak T, Łojko-Dankowska A, Matuszak M, Gil L, Puła B, Szukalski Ł, Końska A, Zaucha JM, Walewski J, Mikulski D, Czabak O, Robak T, Jiang K, Cooperrider JH, Jakubowiak AJ, Derman BA. Mass spectrometry-based assessment of M protein in peripheral blood during maintenance therapy in multiple myeloma. Blood 2024; 144:955-963. [PMID: 38713888 PMCID: PMC11406170 DOI: 10.1182/blood.2024024041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 05/09/2024] Open
Abstract
ABSTRACT Mass spectrometry (MS) can detect multiple myeloma-derived monoclonal proteins in the peripheral blood (PB) with high sensitivity, potentially serving as a PB assay for measurable residual disease (MRD). This study evaluated the significance of PB MS MRD negativity during posttransplant therapy in patients with newly diagnosed multiple myeloma. Serum samples from 138 patients treated in the phase 3 ATLAS trial of posttransplant maintenance with either carfilzomib, lenalidomide, and dexamethasone, or with lenalidomide alone were analyzed using EXENT MS methodology. We established feasibility of measuring MRD by MS in the PB in the posttransplant setting, despite unavailability of pretreatment calibration samples. There was high agreement between MRD by MS in the PB and paired bone marrow (BM) MRD results at the 10-5 threshold, assessed by either next-generation sequencing (NGS) or multiparameter flow cytometry (MFC) (70% and 67%, respectively). Agreement between PB MS and both BM MRD methods was lowest early after transplant and increased with time. MS negativity was associated with improved progression-free survival (PFS), which, in landmark analysis, reached statistical significance after 18 cycles after transplant. Combined PB/BM MRD negativity by MFC or NGS was associated with superior PFS compared with MRD negativity by only 1 modality. Sustained MS negativity carried similar prognostic performance to sustained BM MRD negativity at the 10-5 threshold. Overall, posttransplant MS assessment was feasible and provided additional prognostic information to BM MRD negativity. Further studies are needed to confirm the role and optimal timing of MS in disease evaluation algorithms. The ATLAS trial is registered at www.clinicaltrials.gov as #NCT02659293.
Collapse
Affiliation(s)
- Tadeusz Kubicki
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
- Poznań University of Medical Sciences, Poznań, Poland
| | | | | | | | | | | | | | - Jarosław Czyż
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | | | | | | | | | - Adam Nowicki
- Poznań University of Medical Sciences, Poznań, Poland
| | | | | | | | - Lidia Gil
- Poznań University of Medical Sciences, Poznań, Poland
| | - Bartosz Puła
- Institute of Hematology and Blood Transfusion, Warsaw, Poland
| | - Łukasz Szukalski
- Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
| | | | | | - Jan Walewski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Olga Czabak
- Medical University of Lublin, Lublin, Poland
| | | | - Ken Jiang
- Section of Hematology/Oncology, University of Chicago, Chicago, IL
| | | | | | | |
Collapse
|
30
|
Pasquini MC, Wallace PK, Logan B, Kaur M, Tario JD, Howard A, Zhang Y, Brunstein C, Efebera Y, Geller N, Giralt S, Hari P, Horowitz MM, Koreth J, Krishnan A, Landau H, Somlo G, Shah N, Stadtmauer E, Vogl DT, Vesole DH, McCarthy PL, Hahn T. Minimal Residual Disease Status in Multiple Myeloma 1 Year After Autologous Hematopoietic Cell Transplantation and Lenalidomide Maintenance Are Associated With Long-Term Overall Survival. J Clin Oncol 2024; 42:2757-2768. [PMID: 38701390 PMCID: PMC11634105 DOI: 10.1200/jco.23.00934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 01/31/2024] [Accepted: 03/01/2024] [Indexed: 05/05/2024] Open
Abstract
PURPOSE Prognostic Immunophenotyping in Myeloma Response (PRIMeR) is an ancillary study of minimal residual disease (MRD) assessment for multiple myeloma by next-generation multiparameter flow cytometry (MFC). Patients were enrolled on a three-arm randomized control trial (Blood and Marrow Transplants Clinical Trials Network 0702 Stem Cell Transplant for Myeloma in Combination of Novel Agents [STaMINA]; ClinicalTrials.gov identifier: NCT01109004). METHODS Four hundred and thirty-five patients consented to the MRD panel, which included 10 monoclonal antibodies measured via six-color MFC. MRD was measured at baseline/preautologous hematopoietic cell transplant (BL/preAutoHCT), premaintenance (PM), and 1 year (Y1) after AutoHCT with a sensitivity of 10-5 to 10-6. The primary objective was to assess MRD-negative (MRDneg) at 1 year after AutoHCT and progression-free survival and overall survival (PFS/OS). RESULTS Similar to the STaMINA results, at a median follow-up of 70 months, there was no significant difference in PFS/OS by treatment arm in the PRIMeR patients. MRDneg at all three time points was associated with significantly improved PFS, and MRDneg at Y1 had significantly longer OS. Multivariate analysis of PFS, adjusting for disease risk and treatment arm, demonstrated hazard ratios (HRs) in MRD-positive patients compared with MRDneg patients at BL, PM, and Y1 of 1.55 (P = .0074), 1.83 (P = .0007), and 3.61 (P < .0001), respectively. Corresponding HRs for OS were 1.19 (P = .48), 0.88 (P = .68), and 3.36 (P < .001). Patients with sustained MRDneg or who converted to MRDneg by Y1 had similar PFS/OS. CONCLUSION To our knowledge, this first, prospective US cooperative group, multicenter study demonstrates that MRDneg at Y1 after AutoHCT with lenalidomide maintenance is prognostic for improved 6-year PFS and OS. Serial MRD measurements may direct trials to test how further therapy may improve long-term PFS and OS.
Collapse
Affiliation(s)
| | | | | | | | | | - Alan Howard
- National Marrow Donor Program, Minneapolis, MN
| | - Yali Zhang
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | | | - Yvonne Efebera
- OhioHealth, Columbus, OH
- Ohio State University, Columbus, OH
| | - Nancy Geller
- National Heart, Lung and Blood Institute, Bethesda, MD
| | - Sergio Giralt
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | - Nina Shah
- University California San Francisco, San Francisco, CA
| | | | - Dan T Vogl
- University of Pennsylvania, Philadelphia, PA
| | | | | | - Theresa Hahn
- Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
31
|
Gu X, Tang W, Zhang L, Zheng Y, Pan L, Niu T. Maintenance therapy for cytogenetically high-risk multiple myeloma: landscape in the era of novel drugs. Clin Exp Med 2024; 24:179. [PMID: 39105954 PMCID: PMC11303491 DOI: 10.1007/s10238-024-01445-6] [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: 06/03/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Although the significant strides in novel therapeutic approaches have prolonged the survival of multiple myeloma (MM) patients, the unfavorable prognosis of cytogenetically high-risk newly diagnosed MM (NDMM) remains intractable with the lack of consensus regarding the choice of maintenance regimens. Therefore, this study was initiated with the aim of examining the effectiveness of various maintenance treatments for this group of patients in jeopardy. Overall, 17 studies with 1937 high-risk NDMM patients were included in the network meta-analysis. Combination therapies involving novel drugs presented encouraging prospects in the maintenance phase, while the patients and circumstances for the application of different regimens still needed to be further distinguished and clarified. To investigate the current status of maintenance therapy of high-risk NDMM patients in clinical practice, a real-world cohort of high-risk NDMM was retrospectively incorporated 80 patients with lenalidomide maintenance and 53 patients with bortezomib maintenance, presenting the median PFS of 31.7 months and 30.4 months, respectively (p = 0.874, HR = 0.966, 95% CI: 0.628-1.486). Collectively, this study illuminated the present constraints of conventional approaches during the maintenance phase for high-risk NDMM patients while highlighting the future potential associated with enhanced regimens integrating novel medications.
Collapse
Affiliation(s)
- Xinyuan Gu
- Department of Hematology, Institute of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
- Sichuan University, Chengdu, China
| | - Wenjiao Tang
- Department of Hematology, Institute of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| | - Li Zhang
- Department of Hematology, Institute of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China.
| | - Yuhuan Zheng
- Department of Hematology, Institute of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| | - Ling Pan
- Department of Hematology, Institute of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, Institute of Hematology, West China Hospital, Sichuan University, #37 Guo Xue Xiang Street, Chengdu, 610041, China
| |
Collapse
|
32
|
Bommier C, Maurer MJ, Lambert J. What clinicians should know about surrogate end points in hematologic malignancies. Blood 2024; 144:11-20. [PMID: 38603637 DOI: 10.1182/blood.2023022269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/14/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
ABSTRACT Use of surrogates as primary end points is commonplace in hematology/oncology clinical trials. As opposed to prognostic markers, surrogates are end points that can be measured early and yet can still capture the full effect of treatment, because it would be captured by the true outcome (eg, overall survival). We discuss the level of evidence of the most commonly used end points in hematology and share recommendations on how to apply and evaluate surrogate end points in research and clinical practice. Based on the statistical literature, this clinician-friendly review intends to build a bridge between clinicians and surrogacy specialists.
Collapse
Affiliation(s)
- Côme Bommier
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Epidemiology and Clinical Statistics for Tumor, Respiratory, and Resuscitation Assessments Team, INSERM, U1153, Assistance Publique-Hôpitaux de Paris Hôpital St Louis, Université Paris Cité, Paris, France
| | - Matthew John Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Jerome Lambert
- Epidemiology and Clinical Statistics for Tumor, Respiratory, and Resuscitation Assessments Team, INSERM, U1153, Assistance Publique-Hôpitaux de Paris Hôpital St Louis, Université Paris Cité, Paris, France
| |
Collapse
|
33
|
[Chinese expert consensus on minimal residual disease detection in multiple myeloma based on bone marrow samples (2024)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:534-541. [PMID: 39134483 PMCID: PMC11310804 DOI: 10.3760/cma.j.cn121090-20240430-00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Indexed: 12/06/2024]
Abstract
Multiple myeloma (MM) is the second most common hematologic malignant tumor. Standard holistic treatment model and new drug-based regimens have greatly improved the survival of patients with MM; however, minimal residual disease (MRD) causes relapse in most patients. Therefore, combining MRD testing on the basis of traditional serological efficacy evaluation is necessary to achieve a more accurate assessment of the patient's disease status. At present, next-generation flow cytometry (NGF) and next-generation sequencing (NGS) are the mainstream technologies for detecting MRD based on bone marrow samples. To standardize and normalize MRD detection, the expert group discussed and formulated Chinese expert consensus on the application of NGF and NGS technology for bone marrow MRD detection in patients with MM.
Collapse
|
34
|
Chen J, Gale RP, Hu Y, Yan W, Wang T, Zhang W. Measurable residual disease (MRD)-testing in haematological and solid cancers. Leukemia 2024; 38:1202-1212. [PMID: 38637690 PMCID: PMC11147778 DOI: 10.1038/s41375-024-02252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Affiliation(s)
- Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
35
|
Ishii A, Tsukamoto S, Mimura N, Miyamoto-Nagai Y, Isshiki Y, Matsui S, Nakao S, Shibamiya A, Hino Y, Kayamori K, Oshima-Hasegawa N, Muto T, Takeda Y, Suichi T, Misawa S, Ohwada C, Yokote K, Kuwabara S, Nakaseko C, Takamatsu H, Sakaida E. Detection of clonal plasma cells in POEMS syndrome using multiparameter flow cytometry. Sci Rep 2024; 14:10362. [PMID: 38710832 DOI: 10.1038/s41598-024-61034-1] [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: 04/06/2023] [Accepted: 04/30/2024] [Indexed: 05/08/2024] Open
Abstract
POEMS syndrome (polyneuropathy, organomegaly, endocrinopathy, monoclonal protein [M-protein], and skin changes) is a rare systemic disorder characterized by various symptoms caused by underlying plasma cell (PC) dyscrasia. Detection of monoclonal PCs is mandatory for the diagnosis of POEMS syndrome; however, the usefulness of EuroFlow-based next-generation flow cytometry (EuroFlow-NGF) in POEMS syndrome for detecting monoclonal PCs in bone marrow (BM) and the gating strategy suitable for flow cytometry study of POEMS syndrome remain unknown. We employed EuroFlow-NGF-based single-tube eight-color multiparameter flow cytometry (MM-flow) and established a new gating strategy (POEMS-flow) to detect the monoclonal PCs in POEMS syndrome, gating CD38 broadly from dim to bright and CD45 narrowly from negative to dim compared to MM-flow. MM-flow detected monoclonal PCs in 9/25 (36.0%) cases, including 2/2 immunofixation electrophoresis (IFE)-negative cases (100%). However, POEMS-flow detected monoclonal PCs in 18/25 cases (72.0%), including 2/2 IFE-negative cases (100%). POEMS-flow detected monoclonal PCs with immunophenotypes of CD19- in 17/18 (94.4%). In six cases where post-treatment samples were available, the size of the clones was significantly reduced after the treatment (P = 0.031). POEMS-flow can enhance the identification rate of monoclonal PCs in POEMS syndrome and become a valuable tool for the diagnosis of POEMS syndrome.
Collapse
Affiliation(s)
- Arata Ishii
- Department of Hematology, Chiba University Hospital, Chiba, Japan
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shokichi Tsukamoto
- Department of Hematology, Chiba University Hospital, Chiba, Japan.
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Naoya Mimura
- Department of Hematology, Chiba University Hospital, Chiba, Japan
- Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba, Japan
| | | | - Yusuke Isshiki
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | | | - Sanshiro Nakao
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Asuka Shibamiya
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Yutaro Hino
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Kensuke Kayamori
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | | | - Tomoya Muto
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Yusuke Takeda
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Tomoki Suichi
- Department of Neurology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Sonoko Misawa
- Department of Neurology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Chikako Ohwada
- Department of Hematology, Chiba University Hospital, Chiba, Japan
- Department of Hematology, International University of Health and Welfare, Narita, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Chiaki Nakaseko
- Department of Hematology, International University of Health and Welfare, Narita, Japan
| | - Hiroyuki Takamatsu
- Department of Hematology, Institute of Medical, Pharmaceutical and Health Sciences, Faculty of Medicine, Kanazawa University, Kanazawa, Japan
| | - Emiko Sakaida
- Department of Hematology, Chiba University Hospital, Chiba, Japan
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba, Japan
| |
Collapse
|
36
|
Ludwig H, Melchardt T, Sormann S, Schreder M, Andel J, Hartmann B, Tinchon C, Zojer N, Gunsilius E, Podar K, Egle A, Willenbacher W, Wöll E, Ruckser R, Bozic B, Krauth MT, Petzer A, Schmitt C, Machherndl-Spandl S, Agis H, Fillitz M, Wang SY, Zabernigg A, Knop S, Paiva B, Greil R. Randomized comparison between KTd and KRd induction therapy followed by maintenance therapy with K or observation in transplant-ineligible patients with newly diagnosed multiple myeloma. Am J Hematol 2024; 99:1008-1011. [PMID: 38425185 DOI: 10.1002/ajh.27280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/16/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Randomized comparison between KTd and KRd induction followed by second randomization to carfilzomib in transplant-ineligable patients with newly diagnosed multiple myeloma.
Collapse
Affiliation(s)
- Heinz Ludwig
- Department of Medicine I, Clinic Ottakring, Wilhelminen Cancer Research Institute, Vienna, Austria
| | - Thomas Melchardt
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, Salzburg, Austria
| | - Siegfried Sormann
- Department of Hematology, University Clinic for Internal Medicine, Graz, Austria
| | | | - Johannes Andel
- Department of Internal Medicine II, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr, Austria
| | - Bernd Hartmann
- Department of Internal Medicine II, LKH Rankweil, Rankweil, Austria
| | - Christoph Tinchon
- Department for Hematology, Oncology and Palliative Care, LKH Hochsteiermark, Standort Leoben, Standort Leoben, Austria
| | - Niklas Zojer
- Department of Medicine I, Clinic Ottakring, Vienna, Austria
| | - Eberhard Gunsilius
- Department of Internal Medicine V, Medical University Innsbruck, Innsbruck, Austria
| | - Klaus Podar
- Department of Internal Medicine II, University Hospital Krems, Krems an der Donau, Austria
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Alexander Egle
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, Salzburg, Austria
| | - Wolfgang Willenbacher
- Department of Internal Medicine V, Medical University Innsbruck, Innsbruck, Austria
- syndena GmbH, Connect to Cure, Innsbruck, Austria
| | - Ewald Wöll
- Department of Internal Medicine, St. Vinzenz Krankenhaus Zams, Zams, Austria
| | | | - Boris Bozic
- Department of Medicine II, Clinic Donaustadt, Vienna, Austria
| | - Maria-Theresa Krauth
- University Clinic for Internal Medicine I, AKH, Medical University of Vienna, Vienna, Austria
| | - Andreas Petzer
- Department of Internal Medicine I, BHS Linz, Linz, Austria
| | - Clemens Schmitt
- Clinic for Internal Medicine 3, Kepler University Clinic Linz, Linz, Austria
| | | | - Hermine Agis
- Department of Internal Medicine I, Division of Oncology, Medical University Vienna, Vienna, Austria
| | - Michael Fillitz
- Department of Internal Medicine, Hanusch Krankenhaus, Vienna, Austria
| | - Song-Yau Wang
- Medical Clinic and Policlinic I, University Clinic Leipzig, Leipzig, Germany
| | - August Zabernigg
- Department of Internal Medicine, Kufstein County Hospital, Kufstein, Austria
| | - Stefan Knop
- Klinik für Innere Medizin 5, Schwerpunkt Onkologie/Hämatologie, Klinikum Nürnberg Nord, Nürnberg, Germany
| | - Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), CCUN, IDISNA, Pamplona, Spain
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
37
|
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
|
38
|
Tembhare PR, Sriram H, Khanka T, Gawai S, Bagal B, Ghogale SG, Deshpande N, Girase K, Patil J, Hasan SK, Shetty D, Ghosh K, Chatterjee G, Rajpal S, Patkar NV, Jain H, Punatar S, Gokarn A, Nayak L, Mirgh S, Jindal N, Sengar M, Khattry N, Subramanian PG, Gujral S. Circulating tumor plasma cells and peripheral blood measurable residual disease assessment in multiple myeloma patients not planned for upfront transplant. Hemasphere 2024; 8:e63. [PMID: 38566804 PMCID: PMC10983024 DOI: 10.1002/hem3.63] [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: 11/10/2023] [Revised: 01/16/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Circulating tumor plasma cells (CTPCs) provide a noninvasive alternative for measuring tumor burden in newly diagnosed multiple myeloma (NDMM). Moreover, measurable residual disease (MRD) assessment in peripheral blood (PBMRD) can provide an ideal alternative to bone marrow MRD, which is limited by its painful nature and technical challenges. However, the clinical significance of PBMRD in NDMM still remains uncertain. Additionally, data on CTPC in NDMM patients not treated with transplant are scarce. We prospectively studied CTPC and PBMRD in 141 NDMM patients using highly sensitive multicolor flow cytometry (HS-MFC). PBMRD was monitored at the end of three cycles (PBMRD1) and six cycles (PBMRD2) of chemotherapy in patients with detectable baseline CTPC. Patients received bortezomib-based triplet therapy and were not planned for an upfront transplant. Among baseline risk factors, CTPC ≥ 0.01% was independently associated with poor progression-free survival (PFS) (hazard ratio [HR] = 2.77; p = 0.0047) and overall survival (OS) (HR = 2.9; p = 0.023) on multivariate analysis. In patients with detectable baseline CTPC, undetectable PBMRD at both subsequent time points was associated with longer PFS (HR = 0.46; p = 0.0037), whereas detectable PBMRD at any time point was associated with short OS (HR = 3.25; p = 0.004). Undetectable combined PBMRD (PBMRD1 and PBMRD2) outperformed the serum-immunofixation-based response. On multivariate analysis, detectable PBMRD at any time point was independently associated with poor PFS (HR = 2.0; p = 0.025) and OS (HR = 3.97; p = 0.013). Thus, our findings showed that CTPC and PBMRD assessment using HS-MFC provides a robust, noninvasive biomarker for NDMM patients not planned for an upfront transplant. Sequential PBMRD monitoring has great potential to improve the impact of the existing risk stratification and response assessment models.
Collapse
Affiliation(s)
- Prashant R. Tembhare
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Harshini Sriram
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Twinkle Khanka
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sanghamitra Gawai
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Bhausaheb Bagal
- Department of Medical Oncology, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Sitaram G. Ghogale
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Nilesh Deshpande
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Karishma Girase
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Jagruti Patil
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Syed Khaizer Hasan
- Hasan Laboratory, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Dhanalaxmi Shetty
- Department of Cancer Cytogenetics, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Kinjalka Ghosh
- Department of Biochemistry, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Gaurav Chatterjee
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sweta Rajpal
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Nikhil V. Patkar
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Hasmukh Jain
- Department of Medical Oncology, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Sachin Punatar
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Anant Gokarn
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Lingaraj Nayak
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sumeet Mirgh
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Nishant Jindal
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| | - Navin Khattry
- Bone Marrow Transplant Unit, Department of Medical Oncology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Papagudi G. Subramanian
- Department of Hematopathology, ACTREC, Tata Memorial CentreHBNI UniversityNavi MumbaiMaharashtraIndia
| | - Sumeet Gujral
- Department of Pathology, Tata Memorial Hospital, Tata Memorial CentreHBNI UniversityMumbaiMaharashtraIndia
| |
Collapse
|
39
|
Fan H, Wang B, Shi L, Pan N, Yan W, Xu J, Gong L, Li L, Liu Y, Du C, Cui J, Zhu G, Deng S, Sui W, Xu Y, Yi S, Hao M, Zou D, Chen X, Qiu L, An G. Monitoring Minimal Residual Disease in Patients with Multiple Myeloma by Targeted Tracking Serum M-Protein Using Mass Spectrometry (EasyM). Clin Cancer Res 2024; 30:1131-1142. [PMID: 38170583 PMCID: PMC10940853 DOI: 10.1158/1078-0432.ccr-23-2767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/10/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE We investigated both the clinical utilities and the prognostic impacts of the clonotypic peptide mass spectrometry (MS)-EasyM, a blood-based minimal residual disease (MRD) monitoring protocol in multiple myeloma. EXPERIMENTAL DESIGN A total of 447 sequential serum samples from 56 patients with multiple myeloma were analyzed using EasyM. Patient-specific M-protein peptides were sequenced from diagnostic samples; sequential samples were quantified by EasyM to monitor the M-protein. The performance of EasyM was compared with serum immunofixation electrophoresis (IFE), bone marrow multiparameter flow cytometry (MFC), and next-generation flow cytometry (NGF) detection. The optimal balance of EasyM sensitivity/specificity versus NGF (10-5 sensitivity) was determined and the prognostic impact of MS-MRD status was investigated. RESULTS Of the 447 serum samples detected and measured by EasyM, 397, 126, and 92 had time-matching results for comparison with serum IFE, MFC-MRD, and NGF-MRD, respectively. Using a dotp >0.9 as the MS-MRD positive, sensitivity was 99.6% versus IFE and 100.0% versus MFC and NGF. Using an MS negative cutoff informed by ROC analysis (<1.86% of that at diagnosis), EasyM sensitivity remained high versus IFE (88.3%), MFC (85.1%), and NGF (93.2%), whereas specificity increased to 90.4%, 55.8%, and 93.2%, respectively. In the multivariate analysis, older diagnostic age was an independent predictor for progression-free survival [PFS; high risk (HR), 3.15; 1.26-7.86], the best MS-MRD status (MS-MRD negative) was independent predictor for both PFS (HR, 0.25; 0.12-0.52) and overall survival (HR, 0.16; 0.06-0.40). CONCLUSIONS EasyM is a highly sensitive and minimal invasive method of MRD monitoring in multiple myeloma; MS-MRD had significant predictive ability for survival outcomes.
Collapse
Affiliation(s)
- Huishou Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Bing Wang
- Shanghai Kuaixu Biotechnology Co., Ltd., Shanghai, China
| | - Lihui Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ni Pan
- Shanghai Kuaixu Biotechnology Co., Ltd., Shanghai, China
| | - Wenqiang Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jingyu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lingna Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuntong Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jian Cui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guoqing Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiequn Chen
- Department of Hematology, Affiliated Hospital of Northwest University, Institute of Hematology, Northwest University, Xian, Shaanxi, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
40
|
Partanen A, Waage A, Peceliunas V, Schjesvold F, Anttila P, Säily M, Uttervall K, Putkonen M, Carlson K, Haukas E, Sankelo M, Szatkowski D, Hansson M, Marttila A, Svensson R, Axelsson P, Lauri B, Mikkola M, Karlsson C, Abelsson J, Ahlstrand E, Sikiö A, Klimkowska M, Matuzeviciene R, Fenstad MH, Ilveskero S, Pelliniemi TT, Nahi H, Silvennoinen R. Ixazomib, Lenalidomide, and Dexamethasone (IRD) Treatment with Cytogenetic Risk-Based Maintenance in Transplant-Eligible Myeloma: A Phase 2 Multicenter Study by the Nordic Myeloma Study Group. Cancers (Basel) 2024; 16:1024. [PMID: 38473382 DOI: 10.3390/cancers16051024] [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: 01/27/2024] [Revised: 02/17/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Scarce data exist on double maintenance in transplant-eligible high-risk (HR) newly diagnosed multiple myeloma (NDMM) patients. This prospective phase 2 study enrolled 120 transplant-eligible NDMM patients. The treatment consisted of four cycles of ixazomib-lenalidomide-dexamethasone (IRD) induction plus autologous stem cell transplantation followed by IRD consolidation and cytogenetic risk-based maintenance therapy with lenalidomide + ixazomib (IR) for HR patients and lenalidomide (R) alone for NHR patients. The main endpoint of the study was undetectable minimal residual disease (MRD) with sensitivity of <10-5 by flow cytometry at any time, and other endpoints were progression-free survival (PFS) and overall survival (OS). We present the preplanned analysis after the last patient has been two years on maintenance. At any time during protocol treatment, 28% (34/120) had MRD < 10-5 at least once. At two years on maintenance, 66% of the patients in the HR group and 76% in the NHR group were progression-free (p = 0.395) and 36% (43/120) were CR or better, of which 42% (18/43) had undetectable flow MRD <10-5. Altogether 95% of the patients with sustained MRD <10-5, 82% of the patients who turned MRD-positive, and 61% of those with positive MRD had no disease progression at two years on maintenance (p < 0.001). To conclude, prolonged maintenance with all-oral ixazomib plus lenalidomide might improve PFS in HR patients.
Collapse
Affiliation(s)
- Anu Partanen
- Department of Medicine, Kuopio University Hospital, 70210 Kuopio, Finland
| | - Anders Waage
- Department of Hematology, St. Olavs Hospital, 7030 Trondheim, Norway
| | - Valdas Peceliunas
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital, 08661 Vilnius, Lithuania
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, 0450 Oslo, Norway
- KG Jebsen Center for B Cell Malignancies, University of Oslo, 0316 Oslo, Norway
| | - Pekka Anttila
- Helsinki University Hospital Cancer Center Hematology, University of Helsinki, 00029 Helsinki, Finland
| | - Marjaana Säily
- Hematology-Oncology Unit, Oulu University Hospital Hematology, 90220 Oulu, Finland
| | - Katarina Uttervall
- Medical Unit Hematology, Karolinska University Hospital, 171 64 Solna, Sweden
- Department of Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mervi Putkonen
- Department of Medicine, Turku University Hospital, 20521 Turku, Finland
| | - Kristina Carlson
- Department of Hematology, Uppsala University Hospital, 751 85 Uppsala, Sweden
| | - Einar Haukas
- Stavanger University Hospital, 4011 Stavanger, Norway
| | - Marja Sankelo
- Hematology Unit, Department of Internal Medicine, Tampere University Hospital Hematology, 33520 Tampere, Finland
| | - Damian Szatkowski
- Department of Oncology, Hematology and Palliative Care, Foerde Central Hospital, 6812 Foerde, Norway
| | - Markus Hansson
- Department of Hematology, Skåne University Hospital, 222 42 Lund, Sweden
| | - Anu Marttila
- Department of Medicine, Kymenlaakso Central Hospital, 48210 Kotka, Finland
| | - Ronald Svensson
- Department of Hematology, Linköping University Hospital, 581 85 Linköping, Sweden
| | - Per Axelsson
- Department of Haematology, Helsingborg Hospital, 252 23 Helsingborg, Sweden
| | - Birgitta Lauri
- Department of Hematology, Sunderby Hospital, 971 80 Luleå, Sweden
| | - Maija Mikkola
- Department of Medicine, Päijät-Häme Central Hospital, 15850 Lahti, Finland
| | - Conny Karlsson
- Department of Haematology, Halland Hospital, 302 33 Halmstad, Sweden
| | - Johanna Abelsson
- Department of Hematology, Uddevalla Hospital, 451 53 Uddevalla, Sweden
| | - Erik Ahlstrand
- Department of Medicine, Örebro University Hospital, 701 85 Örebro, Sweden
| | - Anu Sikiö
- Department of Medicine, Central Finland Central Hospital, 40620 Jyväskylä, Finland
| | - Monika Klimkowska
- Department of Clinical Pathology and Cytology, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Reda Matuzeviciene
- Department of Physiology, Biochemistry, Microbiology and Laboratory Medicine, Biomedical Sciences Institute, Vilnius University Hospital and Vilnius University Faculty of Medicine, 03101 Vilnius, Lithuania
| | - Mona Hoysaeter Fenstad
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital, 7030 Trondheim, Norway
| | - Sorella Ilveskero
- Clinical Chemistry, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland
| | | | - Hareth Nahi
- Hematology Centre, Karolinska University Hospital Huddinge, 141 57 Stockholm, Sweden
| | - Raija Silvennoinen
- Helsinki University Hospital Cancer Center Hematology, University of Helsinki, 00029 Helsinki, Finland
| |
Collapse
|
41
|
Wang J, Li J, Zhang R, Li J, Chen L, Jin Y. Real-world prognostic significance of attaining minimal residual disease negativity in newly diagnosed multiple myeloma. Discov Oncol 2024; 15:38. [PMID: 38367151 PMCID: PMC10874347 DOI: 10.1007/s12672-024-00891-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 02/11/2024] [Indexed: 02/19/2024] Open
Abstract
The aim of the study was to evaluate the prognostic impact of minimal residual disease (MRD) in the real-world setting and the interaction between MRD and molecular risk, clinical response and autologous stem-cell transplant (ASCT). A retrospective analysis of 275 newly diagnosed multiple myeloma (NDMM) patients who achieved very good partial remission (VGPR) or better before maintenance were involved. We examined MRD status by multiparameter flow cytometry (MFC). At a median follow-up of 37 months (4-88 months), In patients who achieved ≥ VGPR, those with MRD negativity had significantly longer PFS (51 vs. 26 months; P < 0.001) and OS (Not reached: NR vs. 62 months, P < 0.001) than those with MRD positivity. MRD positivity was the independent prognostic factor for PFS with hazard ratios of 2.650 (95% CI 1.755-4.033, P < 0.001) and OS with hazard ratios of 2.122 (95% CI 1.155-3.899, P = 0.015). Achieving MRD negativity was able to ameliorate a poor prognosis associated with genetic high risk. MRD negativity was associated with better PFS regardless of ASCT treatment. MRD status was more predictable for clinical outcome than conventional clinical responses. Moreover, Sustained MRD negativity ≥ 12 or ≥ 24 months improved both PFS and OS. Patients with NDMM who achieved MRD-negative status or sustained MRD negativity had deep remission and improved clinical outcomes regardless of high-risk cytogenetics, ASCT and clinical responses in a real-world setting.
Collapse
Affiliation(s)
- Jing Wang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Jing Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Run Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, China
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, China.
| | - Yuanyuan Jin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, No. 300 Guangzhou Road, Nanjing, 210029, China.
| |
Collapse
|
42
|
Cui J, Yu T, Lv R, Liu J, Fan H, Yan W, Xu J, Du C, Deng S, Sui W, Ho M, Xu Y, Anderson KC, Dong X, Qiu L, An G. Longitudinal genetically detectable minimal residual disease by fluorescence in situ hybridization confers a poor prognosis in myeloma. Ther Adv Med Oncol 2024; 16:17588359231221340. [PMID: 38249329 PMCID: PMC10799601 DOI: 10.1177/17588359231221340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/29/2023] [Indexed: 01/23/2024] Open
Abstract
Background Deeper depth of response (DpR) after induction therapy, especially gain of negative minimal residual disease (MRD), has been linked to prolonged survival in multiple myeloma (MM). However, flow-MRD examination focuses on the numbers but not on the biological characteristics of residual plasma cells (PCs). Objectives To explore whether the genetic features of residual tumor cells affect the survival time of patients with MM. Design A retrospective cohort study. Methods We investigated the clonality of cytogenetic abnormalities (CAs) of the residual PCs using interphase fluorescence in situ hybridization (iFISH) in the National Longitudinal Cohort of Hematological Diseases in China (NCT04645199). Here, a longitudinal cohort of 269 patients with patient-paired diagnostic and post-induction iFISH results was analyzed. Results Persistent CAs after induction therapy were detected in about half of the patients (118/269, 43%), and patients with undetectable CAs showed significantly improved survival compared with those with genetically detectable MRD [median progression-free survival (mPFS): 59.7 versus 35.7 months, p < 0.001; median overall survival (mOS): 97.1 versus 68.8 months, p = 0.011]. In addition, different patterns of therapy-induced clonal evolution were observed by comparing the clonal structure of residual PCs with paired baseline samples. Patients who maintained at a high risk during follow-up had the worst survival (mPFS: 30.5 months; mOS: 54.4 months), while those who returned to lower risk or had iFISH- at both time points had the best survival (mPFS: 62.0 months, mOS: not reached). Conclusion These findings highlighted the prognostic value of genetic testing in residual tumor cells, which may provide a deep understanding of clonal evolution and guide clinical therapeutic strategies.
Collapse
Affiliation(s)
- Jian Cui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tengteng Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rui Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jiahui Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huishou Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenqiang Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jingyu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Chenxing Du
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Matthew Ho
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Kenneth C. Anderson
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xifeng Dong
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshandao, Heping District, Tianjin 300052, China
| | - Lugui Qiu
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, 288 Nanjing Road, Heping District, Tianjin 300020, China
| | - Gang An
- Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, 288 Nanjing Road, Heping District, Tianjin 300020, China
| |
Collapse
|
43
|
Alnasser SM, Alharbi KS, Almutairy AF, Almutairi SM, Alolayan AM. Autologous Stem Cell Transplant in Hodgkin's and Non-Hodgkin's Lymphoma, Multiple Myeloma, and AL Amyloidosis. Cells 2023; 12:2855. [PMID: 38132175 PMCID: PMC10741865 DOI: 10.3390/cells12242855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Human body cells are stem cell (SC) derivatives originating from bone marrow. Their special characteristics include their capacity to support the formation and self-repair of the cells. Cancer cells multiply uncontrollably and invade healthy tissues, making stem cell transplants a viable option for cancer patients undergoing high-dose chemotherapy (HDC). When chemotherapy is used at very high doses to eradicate all cancer cells from aggressive tumors, blood-forming cells and leukocytes are either completely or partially destroyed. Autologous stem cell transplantation (ASCT) is necessary for patients in those circumstances. The patients who undergo autologous transplants receive their own stem cells (SCs). The transplanted stem cells first come into contact with the bone marrow and then undergo engraftment, before differentiating into blood cells. ASCT is one of the most significant and innovative strategies for treating diseases. Here we focus on the treatment of Hodgkin's lymphoma, non-Hodgkin's lymphoma, multiple myeloma, and AL amyloidosis, using ASCT. This review provides a comprehensive picture of the effectiveness and the safety of ASCT as a therapeutic approach for these diseases, based on the currently available evidence.
Collapse
Affiliation(s)
- Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.F.A.)
| | - Khalid Saad Alharbi
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.F.A.)
| | - Ali F. Almutairy
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.F.A.)
| | | | | |
Collapse
|
44
|
Pérez-Escurza O, Flores-Montero J, Óskarsson JÞ, Sanoja-Flores L, Del Pozo J, Lecrevisse Q, Martín S, Reed ER, Hákonardóttir GK, Harding S, Þorsteinsdóttir S, Rögnvaldsson S, Love TJ, Durie B, Kristinsson SY, Orfao A. Immunophenotypic assessment of clonal plasma cells and B-cells in bone marrow and blood in the diagnostic classification of early stage monoclonal gammopathies: an iSTOPMM study. Blood Cancer J 2023; 13:182. [PMID: 38072838 PMCID: PMC10711003 DOI: 10.1038/s41408-023-00944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Monoclonal gammopathy of undetermined significance (MGUS) is the earliest discernible stage of multiple myeloma (MM) and Waldenström's macroglobulinemia (WM). Early diagnosis of MG may be compromised by the low-level infiltration, undetectable to low-sensitive methodologies. Here, we investigated the prevalence and immunophenotypic profile of clonal (c) plasma cells (PC) and/or cB-lymphocytes in bone marrow (BM) and blood of subjects with a serum M-component from the iSTOPMM program, using high-sensitive next-generation flow cytometry (NGF), and its utility in the diagnostic classification of early-stage MG. We studied 164 paired BM and blood samples from 82 subjects, focusing the analysis on: 55 MGUS, 12 smoldering MM (SMM) and 8 smoldering WM (SWM). cPC were detected in 84% of the BM samples and cB-lymphocytes in 45%, coexisting in 39% of cases. In 29% of patients, the phenotypic features of cPC and/or cB-lymphocytes allowed a more accurate disease classification, including: 19/55 (35%) MGUS, 1/12 (8%) SMM and 2/8 (25%) SWM. Blood samples were informative in 49% of the BM-positive cases. We demonstrated the utility of NGF for a more accurate diagnostic classification of early-stage MG.
Collapse
Affiliation(s)
- Oihane Pérez-Escurza
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | | | - Luzalba Sanoja-Flores
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedicine of Seville, Department of Hematology, University Hospital Virgen del Rocío of the Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Seville, Spain
| | - Julio Del Pozo
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Martín
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Elín Ruth Reed
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | | | - Sigrún Þorsteinsdóttir
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Rigshospitalet, Copenhagen, Denmark
| | - Sæmundur Rögnvaldsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Thorvardur Jon Love
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Brian Durie
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sigurður Yngvi Kristinsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain.
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
45
|
Yu H, Zhang S, Feng X, Gao F. Multiple myeloma following bone metastasis of renal cell carcinoma: a case report. Front Endocrinol (Lausanne) 2023; 14:1206368. [PMID: 38107521 PMCID: PMC10722259 DOI: 10.3389/fendo.2023.1206368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023] Open
Abstract
Background The clinical manifestations of multiple myeloma (MM) and bone metastatic tumor are both systemic bone pain, which is difficult to distinguish from imaging manifestations, leading to misdiagnosis and missed diagnosis. Case summary We reported a man with a unique case whose tumors were MM with bone metastatic tumor of clear cell renal cell carcinoma (CCRCC). Computed tomography (CT) showed multifocal osteolytic bone destruction, while magnetic resonance imaging (MRI) showed multifocal bone marrow infiltration with soft tissue mass. Pathology and immunohistochemistry established the diagnosis of the coexistence of myeloma with bone metastatic tumor of CCRCC in the spine. Immunotherapy and systemic chemotherapy were adopted in the clinic, and vertebral decompression was performed after anemia was corrected. This case with MM and bone metastatic tumor of CCRCC received radiotherapy and immunotherapy and acquired satisfying outcome after 1 year of follow-up. Conclusion It is difficult to differentiate MM and bone metastatic tumor on imaging, especially when there are bone lesions at the same time, which is an easily missed diagnosis and needs to be comprehensively evaluated in combination with functional procedures, clinical laboratory tests, and histopathology.
Collapse
Affiliation(s)
- Hong Yu
- Department of Radiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shengnan Zhang
- Department of Radiology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaohui Feng
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Gao
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
46
|
Óskarsson JÞ, Rögnvaldsson S, Thorsteinsdottir S, Aspelund T, Gunnarsson SB, Hákonardóttir GK, Sigurðardóttir GÁ, Þórðardóttir ÁR, Gíslason GK, Ólafsson A, Sigurðsson JK, Eyþórsson E, Jónsson Á, Viðarsson B, Önundarson PT, Agnarsson BA, Pálmason R, Sigurðardóttir M, Þorsteinsdóttir I, Ólafsson Í, Harding S, Flores-Montero J, Orfao A, Durie BGM, Love TJ, Kristinsson SY. Determining hemodilution in diagnostic bone marrow aspirated samples in plasma cell disorders by next-generation flow cytometry: Proposal for a bone marrow quality index. Blood Cancer J 2023; 13:177. [PMID: 38040702 PMCID: PMC10692231 DOI: 10.1038/s41408-023-00951-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023] Open
Abstract
Hemodilution of bone marrow (BM) aspirates is a limitation of multiparameter flow cytometry (MFC) in plasma cell disorders. There is a need for a validated approach for assessing sample quality and the distribution of non-plasma cell BM populations by MFC could provide a solution. We evaluated BM-associated cell populations, assessed by next-generation flow cytometry (NGF) and white blood cell (WBC) count in 351 BM aspirated samples from 219 participants with plasma cell disorders in the Iceland Screens, Treats, or Prevents MM study (iStopMM), as markers of hemodilution by their discriminatory ability between first and (generally more hemodiluted) second pull BM aspirated samples. The most discriminating markers were used to derive a novel BM quality index (BMQI). Nucleated red blood cells and myeloid precursors provided the greatest discriminatory ability between first vs second pull samples (area under the curve (AUC): 0.87 and 0.85, respectively), significantly better than B cell precursors (AUC = 0.64; p < 0.001), mast cells (AUC = 0.65; p < 0.001), and the BM WBC count (AUC = 0.77; p < 0.05). We generated a novel BMQI that is intrinsic to current NGF protocols, for evaluating quality of diagnostic BM samples and suggest the use of a BMQI scoring system for interpreting results and guiding appropriate actions.
Collapse
Affiliation(s)
| | - Sæmundur Rögnvaldsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| | - Sigrun Thorsteinsdottir
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Thor Aspelund
- Public Health Sciences, University of Iceland, Reykjavík, Iceland
| | | | | | | | | | | | - Andri Ólafsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | | | | | | | | | | | - Róbert Pálmason
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | | | | | | | | | - Juan Flores-Montero
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca; Biomedical Research Institute of Salamanca (IBSAL) and CIBERONC, Salamanca, Spain
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL-CSIC), Department of Medicine and Cytometry Service (NUCLEUS), University of Salamanca; Biomedical Research Institute of Salamanca (IBSAL) and CIBERONC, Salamanca, Spain
| | - Brian G M Durie
- Cedars-Sinai Samuel Oschin Cancer Center, Los Angeles, CA, USA
| | - Thorvardur Jon Love
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| | - Sigurdur Yngvi Kristinsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Landspítali University Hospital, Reykjavík, Iceland
| |
Collapse
|
47
|
Jin X, Jiang X, Wang W, Liu S, Han B, Han J, Zhuang J. Pure red cell aplasia and minimal residual disease conversion associated with immune reconstitution in a patient with high-risk multiple myeloma. Chronic Dis Transl Med 2023; 9:341-344. [PMID: 37915388 PMCID: PMC10617308 DOI: 10.1002/cdt3.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 11/03/2023] Open
Abstract
A second bone marrow aspiration and biopsy showed pure red cell aplasia in this case.
Collapse
Affiliation(s)
- Xianghong Jin
- Department of HematologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
- Peking Union Medical College, Chinese Academy and Medical SciencesBeijingChina
| | - Xianyong Jiang
- Department of HematologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Wei Wang
- Department of HematologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Shuangjiao Liu
- Department of HematologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Bing Han
- Department of HematologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Jianhua Han
- Department of Clinical LaboratoryPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| | - Junling Zhuang
- Department of HematologyPeking Union Medical College Hospital, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
48
|
Cárdenas MC, García-Sanz R, Puig N, Pérez-Surribas D, Flores-Montero J, Ortiz-Espejo M, de la Rubia J, Cruz-Iglesias E. Recommendations for the study of monoclonal gammopathies in the clinical laboratory. A consensus of the Spanish Society of Laboratory Medicine and the Spanish Society of Hematology and Hemotherapy. Part I: Update on laboratory tests for the study of monoclonal gammopathies. Clin Chem Lab Med 2023; 61:2115-2130. [PMID: 37477188 DOI: 10.1515/cclm-2023-0326] [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: 03/30/2023] [Accepted: 05/29/2023] [Indexed: 07/22/2023]
Abstract
Monoclonal gammopathies (MG) are characterized by the proliferation of plasma cells that produce identical abnormal immunoglobulins (intact or some of their subunits). This abnormal immunoglobulin component is called monoclonal protein (M-protein), and is considered a biomarker of proliferative activity. The identification, characterization and measurement of M-protein is essential for the management of MG. We conducted a systematic review of the different tests and measurement methods used in the clinical laboratory for the study of M-protein in serum and urine, the biochemistry and hematology tests necessary for clinical evaluation, and studies in bone marrow, peripheral blood and other tissues. This review included literature published between 2009 and 2022. The paper discusses the main methodological characteristics and limitations, as well as the purpose and clinical value of the different tests used in the diagnosis, prognosis, monitoring and assessment of treatment response in MG. Included are methods for the study of M-protein, namely electrophoresis, measurement of immunoglobulin levels, serum free light chains, immunoglobulin heavy chain/light chain pairs, and mass spectrometry, and for the bone marrow examination, morphological analysis, cytogenetics, molecular techniques, and multiparameter flow cytometry.
Collapse
Affiliation(s)
- María C Cárdenas
- Department of Clinical Analysis, Hospital Clinico San Carlos, Madrid, Spain
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca, Research Biomedical Institute of Salamanca (IBSAL), CIBERONC and Center for Cancer Research-IBMCC (University of Salamanca-CSIC), Salamanca, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - Noemí Puig
- Hematology Department, University Hospital of Salamanca, Research Biomedical Institute of Salamanca (IBSAL), CIBERONC and Center for Cancer Research-IBMCC (University of Salamanca-CSIC), Salamanca, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - David Pérez-Surribas
- Laboratori Pasteur, Andorra La Vella, Andorra
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| | - Juan Flores-Montero
- Hematology Department, University Hospital of Salamanca, Research Biomedical Institute of Salamanca (IBSAL), CIBERONC and Center for Cancer Research-IBMCC (University of Salamanca-CSIC), Salamanca, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - María Ortiz-Espejo
- Department of Clinical Analysis, Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| | - Javier de la Rubia
- Hematology Department, Hospital Universitario y Politécnico La Fe & Universidad Católica de Valencia, Instituto de Investigación Sanitaria La Fe Centro de Investigación Biomédica en Red de Cáncer, CIBERONC CB16/12/00284, Instituto de Salud Carlos III, Valencia, Spain
- Spanish Society of Hematology and Hemotherapy (SEHH), Madrid, Spain
| | - Elena Cruz-Iglesias
- Department of Laboratory Medicine, Osakidetza Basque Health Service, Basurto University Hospital, Bilbao, Spain
- Protein Commission, Spanish Society of Laboratory Medicine (SEQCML), Barcelona, Spain
| |
Collapse
|
49
|
Mohty M, Avet-Loiseau H, Malard F, Harousseau JL. Potential future direction of measurable residual disease evaluation in multiple myeloma. Blood 2023; 142:1509-1517. [PMID: 37471603 DOI: 10.1182/blood.2023020284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023] Open
Abstract
Multiple myeloma remains an incurable disease plagued by high relapse rates. Deeper and more sustainable responses, however, have been consistently shown to improve outcomes and could eventually pave the way to achieving a cure. Our understanding of disease response has surpassed complete response (CR), because the current definitions are suboptimal, and the treatment goal should aim even beyond stringent CR, toward molecular and flow CR, that is, measurable residual disease (MRD) negativity. It has been more than 20 years since the discrepancy in the outcome between patients in CR with and without MRD has been demonstrated, and the field has come a long way from multiparameter flow cytometry to next-generation flow and next-generation sequencing, able to detect up to a limit of detection of a single myeloma cell from 1 million healthy counterparts. This review aims to summarize the current available data regarding MRD but also its potential future use as a coprimary outcome both in clinical and trial settings as a survival surrogate as well as its use to evaluate treatment efficacy and for adaptive response-based and early-rescue therapy. Furthermore, we discuss whether these concepts are applicable in different settings (eg, newly diagnosed and relapsed/refractory myeloma, patients who are eligible and ineligible for tansplant, and standard- and high-risk disease).
Collapse
Affiliation(s)
- Mohamad Mohty
- UMRS 938, Saint-Antoine Research Center, INSERM, Sorbonne University, Paris, France
- Clinical Hematology and Cellular Therapy Department, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Hervé Avet-Loiseau
- Cancer Research Center of Toulouse, INSERM, Myeloma Genomics Lab University Cancer Institute Toulouse Oncopole, Toulouse, France
- Université Paul Sabatier, Toulouse, France
| | - Florent Malard
- UMRS 938, Saint-Antoine Research Center, INSERM, Sorbonne University, Paris, France
- Clinical Hematology and Cellular Therapy Department, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Luc Harousseau
- Centre René Gauducheau, Institut de Cancérologie de l'Ouest, Nantes-St Herblain, France
| |
Collapse
|
50
|
Feng Y, Qi S, Liu X, Zhang L, Hu Y, Shen Q, Gong X, Zhang W, Wang J, Yan W, Wang T, Wang H, Song Z, Zhu X, Gale RP, Chen J. Have we been qualifying measurable residual disease correctly? Leukemia 2023; 37:2168-2172. [PMID: 37704711 PMCID: PMC10624632 DOI: 10.1038/s41375-023-02026-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Affiliation(s)
- Yahui Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Saibing Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xueou Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Li Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Qiujin Shen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaowen Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Junxia Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wen Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tiantian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huijun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhen Song
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College of Science, Technology and Medicine, London, UK
| | - Junren Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
- Tianjin Institutes of Health Science, Tianjin, China.
| |
Collapse
|