1
|
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] [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
|
2
|
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
|
3
|
Zhou M, Chen Y, Gong Y, Zhu M, Cen J, Pan J, Yan L, Shang J, Jin S, Shi X, Yao W, Yan S, Wu D, Chen S, Fu C, Yao L. Evaluation of next-generation sequencing versus next-generation flow cytometry for minimal-residual-disease detection in Chinese patients with multiple myeloma. Discov Oncol 2024; 15:78. [PMID: 38502423 PMCID: PMC10951185 DOI: 10.1007/s12672-024-00938-w] [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: 01/04/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
PURPOSE To evaluate the efficacy of next-generation sequencing (NGS) in minimal-residual-disease (MRD) monitoring in Chinese patients with multiple myeloma (MM). METHODS This study analyzed 60 Chinese MM patients. During MRD monitoring in these patients' post-therapy, clonal immunoglobulin heavy chain (IGH) rearrangements were detected via NGS using LymphoTrack assays. MRD monitoring was performed using NGS or next-generation flow cytometry (NGF), and the results were compared. Additionally, the sensitivity and reproducibility of the NGS method were assessed. RESULTS The MRD detection range of the NGS method was 10-6-10-1, which suggested good linearity, with a Pearson correlation coefficient of 0.985 and a limit of detection of 10-6. Intra- and inter-assay reproducibility analyses showed that NGS exhibited 100% reproducibility with low variability in clonal cells. At diagnosis, unique clones were found in 42 patients (70.0%) with clonal IGH rearrangements, which were used as clonality markers for MRD monitoring post-therapy. Comparison of NGS and NGF for MRD monitoring showed 79.1% concordance. No samples that tested MRD-positive via NGF were found negative via NGS, indicating the higher sensitivity of NGS. MRD could be detected using NGS in 6 of 7 samples before autologous hematopoietic stem-cell transplantation, and 5 of them tested negative post-transplantation. In contrast, the NGF method could detect MRD in only 1 sample pre-transplantation. CONCLUSION Compared with NGF, NGS exhibits higher sensitivity and reproducibility in MRD detection and can be an effective strategy for MRD monitoring in Chinese MM patients.
Collapse
Affiliation(s)
- Mo Zhou
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
- Hematology Department, Yancheng Third People's Hospital, Yancheng, People's Republic of China
| | - Yan Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Yanlei Gong
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Mingqing Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Jiannong Cen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Jinlan Pan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Lingzhi Yan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Jingjing Shang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Song Jin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Xiaolan Shi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Weiqin Yao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Shuang Yan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People's Republic of China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People's Republic of China
| | - Chengcheng Fu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China
| | - Li Yao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Soochow University, 188 Shizi Street, Suzhou, 215006, People's Republic of China.
| |
Collapse
|
4
|
Guerrero C, Puig N, Cedena MT, Calasanz MJ, Gutierrez NC, Fernandez M, Oriol A, Ríos-Tamayo R, Hernandez MT, Martínez-Martínez R, Bargay J, de Arriba F, Palomera L, Gonzalez-Rodriguez AP, Gonzalez Perez MS, Orfao A, Mateos MV, Martinez-Lopez J, Rosiñol L, Bladé J, Lahuerta JJ, San-Miguel JF, Paiva B. Predictors of unsustained measurable residual disease negativity in transplant-eligible patients with multiple myeloma. Blood 2024; 143:597-603. [PMID: 38048552 DOI: 10.1182/blood.2023022083] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
ABSTRACT The role of measurable residual disease (MRD) negativity as a biomarker to stop treatment is being investigated in transplant-eligible patients with multiple myeloma (MM). Thus, it is important to identify risk factors of MRD resurgence and/or progressive disease (PD) among patients achieving undetectable MRD to avoid undertreating them. Here, we studied 267 newly diagnosed transplant-eligible patients with MM enrolled in the GEM2012MENOS65 and GEM2014MAIN clinical trials who achieved MRD negativity by next-generation flow cytometry. After a median follow-up of 73 months since the first MRD negative assessment, 111 of the 267 (42%) patients showed MRD resurgence and/or PD. The only prognostic factors at diagnosis that predicted MRD resurgence and/or PD were an International Staging System (ISS) 3 and the presence of ≥0.01% circulating tumor cells (CTCs). Failure to achieve MRD negativity after induction also predicted higher risk of MRD resurgence and/or PD. Patients having 0 vs 1 vs ≥2 risk factors (ISS 3, ≥0.01% CTCs, and late MRD negativity) showed 5-year rates of MRD resurgence and/or PD of 16%, 33%, and 57%, respectively (P < .001). Thus, these easily measurable risk factors could help refine the selection of patients for whom treatment cessation after MRD negativity is being investigated in clinical trials. This trial was registered at www.clinicaltrials.gov as NCT01916252 and NCT02406144.
Collapse
Affiliation(s)
- Camila Guerrero
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Cáncer (CIBER-ONC) number CB16/12/00369, Pamplona, Spain
| | - Noemi Puig
- Department of Hematology, Hospital Universitario de Salamanca Hematología, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - María-Teresa Cedena
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María-José Calasanz
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Cáncer (CIBER-ONC) number CB16/12/00369, Pamplona, Spain
| | - Norma C Gutierrez
- Department of Hematology, Hospital Universitario de Salamanca Hematología, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Manuela Fernandez
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Albert Oriol
- Department of Hematology, Institut Català d'Oncologia L'Hospitalet, Barcelona, Spain
| | - Rafael Ríos-Tamayo
- Department of Hematology, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain
| | | | | | - Joan Bargay
- Department of Hematology, Hospital Universitario Son Llatzer, Institut d'Investigacio Illes Balears, Palma de Mallorca, Spain
| | - Felipe de Arriba
- Department of Hematology, Hospital Morales Meseguer, Instituto Murciano de Investigación Biosanitaria-Pascual Parrilla, Universidad de Murcia, Murcia, Spain
| | - Luis Palomera
- Department of Hematology, Hospital Clínico Lozano Blesa, Zaragoza, Spain
| | | | - Marta-Sonia Gonzalez Perez
- Department of Hematology, Complejo Hospitalario Universitario de Santiago de Compostela, Servizio Galego de Saúde, Compostela, Spain
| | - Alberto Orfao
- Department of Hematology, Hospital Universitario de Salamanca Hematología, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - María-Victoria Mateos
- Department of Hematology, Hospital Universitario de Salamanca Hematología, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | | | - Laura Rosiñol
- Department of Hematology, Hospital Clínic, Institut de Recerca Biomedica August Pi i Suñer, Barcelona, Spain
| | - Joan Bladé
- Department of Hematology, Hospital Clínic, Institut de Recerca Biomedica August Pi i Suñer, Barcelona, Spain
| | - Juan-Jose Lahuerta
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Jesus F San-Miguel
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Cáncer (CIBER-ONC) number CB16/12/00369, Pamplona, Spain
| | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Centro de Investigación Biomédica en Red de Cáncer (CIBER-ONC) number CB16/12/00369, Pamplona, Spain
| |
Collapse
|
5
|
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
|
6
|
Kubicki T, Derman BA, Dytfeld D, Jakubowiak AJ. Measurable residual disease in peripheral blood in myeloma: dream or reality. Curr Opin Oncol 2023; 35:574-580. [PMID: 37621165 DOI: 10.1097/cco.0000000000000987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
PURPOSE OF REVIEW Therapeutic advancements in multiple myeloma have led to increasingly deeper and more durable responses, creating a need for highly sensitive and applicable techniques for measurable residual disease (MRD) assessment. Bone marrow assays can deeply assess for MRD, but it is not conducive to performing frequent and dynamic evaluations, which may be needed for MRD-adapted treatment approaches. Recently, numerous techniques for MRD assessment in peripheral blood have come under investigation, and their integration into routine clinical practice is eagerly anticipated. RECENT FINDINGS The identification of circulating tumor cells (CTCs), evaluation of cell-free DNA, and measuring monoclonal protein concentration with mass spectrometry are promising research areas for assessing myeloma in peripheral blood. CTCs assessment and cell-free DNA may carry prognostic significance, but they lack the sensitivity of bone marrow-based techniques. Mass spectrometry has already been implemented in clinical practice in certain centers, but its full potential has yet to be fully realized. This review focuses on recent developments in these fields, emphasizing the potential future roles of these assessments. SUMMARY MRD assessment in peripheral blood is still in the development stage but holds promise for not only complementing bone marrow based evaluations but also potential for improving sensitivity.
Collapse
|
7
|
Yu Z, Xie L, Zhang J, Lin H, Niu T. The evolution of minimal residual disease: key insights based on a bibliometric visualization analysis from 2002 to 2022. Front Oncol 2023; 13:1186198. [PMID: 37534257 PMCID: PMC10391156 DOI: 10.3389/fonc.2023.1186198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/15/2023] [Indexed: 08/04/2023] Open
Abstract
Background The topic of minimal residual disease (MRD) has emerged as a crucial subject matter in the domain of oncology in recent years. The detection and monitoring of MRD have become essential for the diagnosis, treatment, and prognosis of various types of malignancy. Aims The purpose of this study is to explore the research trends, hotspots, and frontiers of MRD in the last two decades through bibliometric analysis. Methods We employed Web of Science databases to carry out a bibliometric visualization analysis of research on 8,913 academic papers about MRD research from 2002 to 2022. VOSviewer, CiteSpace, RStudio, and a bibliometric online analysis platform were mainly used to conduct co-occurrence analysis and cooperative relationship analysis of countries/regions, institutions, journals, and authors in the literature. Furthermore, co-occurrence, co-citation, and burst analyses of keyword and reference were also conducted to generate relevant knowledge maps. Results In the past 20 years, the number of MRD research papers has presented an overall rising trend, going through three stages: a plateau, development, and an explosion. The output of articles in the United States was notably superior and plays a dominant role in this field, and the Netherlands had the highest average citation per article. The most productive and influential institution was the University of Texas MD Anderson Cancer Center. Blood published the most papers and was the most cited journal. A collection of leading academics has come to the fore in the research field, the most prolific of which is Kantarjian HM. It was found that the application of MRD in "acute myeloid leukemia", "acute lymphoblastic leukemia", "multiple myeloma", as well as the detection technology of MRD, are the research hotspots and frontiers in this domain. Furthermore, we analyzed the co-citation network of references and found that the top 10 co-cited references were all associated with MRD in hematological malignancies. Conclusion This bibliometric visualization analysis conducted a thorough exploration into the research hotspots and trends in MRD from 2002 to 2022. Our findings can aid researchers in recognizing possible collaborations, guiding future research directions, and fostering the growth of MRD detection and monitoring technologies.
Collapse
Affiliation(s)
- Zhengyu Yu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Xie
- State Key Laboratory of Wildlife Quarantine and Surveillance (Sichuan), Technology Center of Chengdu Customs, Chengdu, China
| | - Jing Zhang
- State Key Laboratory of Wildlife Quarantine and Surveillance (Sichuan), Technology Center of Chengdu Customs, Chengdu, China
| | - Hua Lin
- State Key Laboratory of Wildlife Quarantine and Surveillance (Sichuan), Technology Center of Chengdu Customs, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
8
|
Lejeune M, Köse MC, Jassin M, Gou MJ, Herbet A, Duray E, Cobraiville G, Foguenne J, Boquet D, Gothot A, Beguin Y, Fillet M, Caers J. Integrative Analysis of Proteomics and Transcriptomics Reveals Endothelin Receptor B as Novel Single Target and Identifies New Combinatorial Targets for Multiple Myeloma. Hemasphere 2023; 7:e901. [PMID: 37359190 PMCID: PMC10289631 DOI: 10.1097/hs9.0000000000000901] [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: 12/22/2022] [Accepted: 04/25/2023] [Indexed: 06/28/2023] Open
Abstract
Despite the recent introduction of next-generation immunotherapeutic agents, multiple myeloma (MM) remains incurable. New strategies targeting MM-specific antigens may result in a more effective therapy by preventing antigen escape, clonal evolution, and tumor resistance. In this work, we adapted an algorithm that integrates proteomic and transcriptomic results of myeloma cells to identify new antigens and possible antigen combinations. We performed cell surface proteomics on 6 myeloma cell lines based and combined these results with gene expression studies. Our algorithm identified 209 overexpressed surface proteins from which 23 proteins could be selected for combinatorial pairing. Flow cytometry analysis of 20 primary samples confirmed the expression of FCRL5, BCMA, and ICAM2 in all samples and IL6R, endothelin receptor B (ETB), and SLCO5A1 in >60% of myeloma cases. Analyzing possible combinations, we found 6 combinatorial pairs that can target myeloma cells and avoid toxicity on other organs. In addition, our studies identified ETB as a tumor-associated antigen that is overexpressed on myeloma cells. This antigen can be targeted with a new monoclonal antibody RB49 that recognizes an epitope located in a region that becomes highly accessible after activation of ETB by its ligand. In conclusion, our algorithm identified several candidate antigens that can be used for either single-antigen targeting approaches or for combinatorial targeting in new immunotherapeutic approaches in MM.
Collapse
Affiliation(s)
- Margaux Lejeune
- Laboratory of Hematology, GIGA I3, University of Liège, Belgium
| | - Murat Cem Köse
- Laboratory of Hematology, GIGA I3, University of Liège, Belgium
| | - Mégane Jassin
- Laboratory of Hematology, GIGA I3, University of Liège, Belgium
| | - Marie-Jia Gou
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Belgium
| | - Amaury Herbet
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Gif-sur-Yvette, France
| | - Elodie Duray
- Laboratory of Hematology, GIGA I3, University of Liège, Belgium
| | - Gaël Cobraiville
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Belgium
| | - Jacques Foguenne
- Department of Hematobiology and Immunohematology, CHU de Liège, Belgium
| | - Didier Boquet
- Université Paris-Saclay, CEA, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Gif-sur-Yvette, France
| | - André Gothot
- Department of Hematobiology and Immunohematology, CHU de Liège, Belgium
| | - Yves Beguin
- Laboratory of Hematology, GIGA I3, University of Liège, Belgium
- Department of Hematology, CHU de Liège, Belgium
| | - Marianne Fillet
- Laboratory for the Analysis of Medicines, Center for Interdisciplinary Research on Medicines (CIRM), University of Liège, Belgium
| | - Jo Caers
- Laboratory of Hematology, GIGA I3, University of Liège, Belgium
- Department of Hematology, CHU de Liège, Belgium
| |
Collapse
|
9
|
Hillengass J, Martin T, Puig N, Paiva B, Usmani S, Kumar S, San-Miguel J. Disease Monitoring In Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:244-248. [PMID: 36774297 DOI: 10.1016/j.clml.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/16/2023]
Affiliation(s)
| | - Tom Martin
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Noemi Puig
- Department of Hematology Salamanca, Salamanca, Spain
| | - Bruno Paiva
- Clínica Universidad de Navarra, CCUN, CIMA, CIBERONC, IDISNA, Pamplona, Spain
| | - Saad Usmani
- Levine Cancer Institute-Atrium Health, Charlotte, NC
| | - Shaji Kumar
- Department of Hematology, Mayo Clinic, Rochester, MN
| | - Jesus San-Miguel
- Clínica Universidad de Navarra, CCUN, CIMA, CIBERONC, IDISNA, Pamplona, Spain.
| |
Collapse
|
10
|
Fitzpatrick MJ, Murali MR, Nardi V. Molecular Diagnostics of Plasma Cell Neoplasms. Surg Pathol Clin 2023; 16:401-410. [PMID: 37149365 DOI: 10.1016/j.path.2023.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Genetic characterization of myeloma at diagnosis by interphase fluorescence in situ hybridization and next-generation sequencing (NGS) can assist with risk stratification and treatment planning. Measurable residual disease (MRD) status after treatment, as evaluated by next-generation flow cytometry or NGS on bone marrow aspirate material, is one of the most important predictors of prognosis. Less-invasive tools for MRD assessment such as liquid biopsy approaches have also recently emerged as potential alternatives.
Collapse
|
11
|
Foureau DM, Paul BA, Guo F, Lipford EH, Fesenkova K, Tjaden E, Drummond K, Bhutani M, Atrash S, Ndiaye A, Varga C, Voorhees PM, Usmani SZ. Standardizing Clinical Workflow for Assessing Minimal Residual Disease by Flow Cytometry in Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:e41-e50. [PMID: 36443182 PMCID: PMC10448729 DOI: 10.1016/j.clml.2022.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Minimal residual disease (MRD) status is an established prognostic biomarker for patients with multiple myeloma. Commonly used MRD testing techniques such as next generation sequencing or next generation flow cytometry can detect as little as one or two multiple myeloma plasma cells in 106 normal bone marrow cells. Early pull of bone marrow aspirates (BMA), necessary to achieve such level of sensitivity, can be difficult to secure in routine clinical practice due to the competing need for early pull samples for clinical response assessment, therefore introducing the risk of analytical interference during MRD testing. METHODS To overcome this challenge, we standardized our workflow for collecting specimens by using a technical first pull after needle repositioning for MRD testing. To capture a comprehensive picture of MRD assay performance and specimen adequacy, we tested for MRD on 556 technical first pull bone marrow aspirates by next generation flow cytometry. Among the specimens, several key multiple myeloma treatment milestones were represented: end of induction therapy, two to three months post-autologous stem cell transplant, early and late stages of maintenance therapy. RESULTS By using the technical first pull bone marrow aspirate, we achieved an analytical assay input of 10 million nucleated cells for 97.5% of specimens. Our analytical sensitivity reached 10-6; (i.e., 10 multiple myeloma plasma cells in 10 × 106 bone marrow cells). Twenty-four percent of specimens were significantly hemodiluted. Low assay input or hemodilution quantifiably lowered the assay sensitivity. CONCLUSION Specimen adequacy is, therefore, an important metric to incorporate into MRD status reporting.
Collapse
Affiliation(s)
- David M Foureau
- Immune Monitoring Core Laboratory, Levine Cancer Institute, Atrium Health, Charlotte, NC.
| | - Barry A Paul
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Fei Guo
- Immune Monitoring Core Laboratory, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Edward H Lipford
- Department of Hematologic Pathology, Atrium Health, Charlotte, NC
| | - Kateryna Fesenkova
- Immune Monitoring Core Laboratory, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Elise Tjaden
- Department of Cancer Biostatistics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Kendra Drummond
- Department of Cancer Biostatistics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Manisha Bhutani
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Shebli Atrash
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Ami Ndiaye
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Cindy Varga
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Peter M Voorhees
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Saad Z Usmani
- Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
12
|
Bergantim R, Peixoto da Silva S, Polónia B, Barbosa MAG, Albergaria A, Lima J, Caires HR, Guimarães JE, Vasconcelos MH. Detection of Measurable Residual Disease Biomarkers in Extracellular Vesicles from Liquid Biopsies of Multiple Myeloma Patients-A Proof of Concept. Int J Mol Sci 2022; 23:13686. [PMID: 36430163 PMCID: PMC9690807 DOI: 10.3390/ijms232213686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
Monitoring measurable residual disease (MRD) is crucial to assess treatment response in Multiple Myeloma (MM). Detection of MRD in peripheral blood (PB) by exploring Extracellular Vesicles (EVs), and their cargo, would allow frequent and minimally invasive monitoring of MM. This work aims to detect biomarkers of MRD in EVs isolated from MM patient samples at diagnosis and remission and compare the MRD-associated content between BM and PB EVs. EVs were isolated by size-exclusion chromatography, concentrated by ultrafiltration, and characterized according to their size and concentration, morphology, protein concentration, and the presence of EV-associated protein markers. EVs from healthy blood donors were used as controls. It was possible to isolate EVs from PB and BM carrying MM markers. Diagnostic samples had different levels of MM markers between PB and BM paired samples, but no differences between PB and BM were found at remission. EVs concentration was lower in the PB of healthy controls than of patients, and MM markers were mostly not detected in EVs from controls. This study pinpoints the potential of PB EVs from MM remission patients as a source of MM biomarkers and as a non-invasive approach for monitoring MRD.
Collapse
Affiliation(s)
- Rui Bergantim
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital Center of São João, 4200-319 Porto, Portugal
- Clinical Hematology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Sara Peixoto da Silva
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Bárbara Polónia
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Mélanie A. G. Barbosa
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - André Albergaria
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Research Innovation Unit, Translational Research & Industry Partnerships Office, i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Jorge Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Research Innovation Unit, Translational Research & Industry Partnerships Office, i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
| | - Hugo R. Caires
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital Center of São João, 4200-319 Porto, Portugal
- Clinical Hematology, FMUP—Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário IUCSESPU, 4585-116 Gandra-Paredes, Portugal
| | - M. Helena Vasconcelos
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
13
|
Dual Negativity of CD56 and CD117 Links to Unfavorable Cytogenetic Abnormalities and Predicts Poor Prognosis in Multiple Myeloma. J Clin Med 2022; 11:jcm11216524. [DOI: 10.3390/jcm11216524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
The prognostic value of CD56 and CD117 expression on myeloma cells is controversial. This study aims to analyze the correlation of CD56 and CD117 expression with cytogenetic abnormalities and survival. A total of 128 patients with newly diagnosed multiple myeloma (NDMM) were recruited in this single-center retrospective study. Flow cytometry and FISH tests of marrow cells were performed for all of the subjects. The statistical methods included a chi-squared test, univariate and multivariate COX regressions, and a Kaplan-Meier survival curve analysis. Regarding the cytogenetics, the incidence of IgH/FGFR3 translocation was more frequent in patients with a negative CD56 (p = 0.003). CD56 negativity was an independent adverse factor associated with a poor prognosis (p = 0.019) and indicated a shorter overall survival (OS) (p = 0.021). Patients with dual negative CD56 and CD117 trended toward a poorer OS (CD56−CD117− vs. CD56+CD117−, p = 0.011; CD56−CD117− vs. CD56+CD117+, p = 0.013). In conclusion, CD56 is a prognostic marker that independently affects OS and is associated with adverse cytogenetic abnormalities. Patients with a dual negativity of CD56 and CD117 have a worse clinical outcome.
Collapse
|
14
|
Vigliotta I, Armuzzi S, Barone M, Solli V, Pistis I, Borsi E, Taurisano B, Mazzocchetti G, Martello M, Poletti A, Sartor C, Rizzello I, Pantani L, Tacchetti P, Papayannidis C, Mancuso K, Rocchi S, Zamagni E, Curti A, Arpinati M, Cavo M, Terragna C. The ALLgorithMM: How to define the hemodilution of bone marrow samples in lymphoproliferative diseases. Front Oncol 2022; 12:1001048. [PMID: 36276072 PMCID: PMC9582597 DOI: 10.3389/fonc.2022.1001048] [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: 07/22/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Minimal residual disease (MRD) is commonly assessed in bone marrow (BM) aspirate. However, sample quality can impair the MRD measurement, leading to underestimated residual cells and to false negative results. To define a reliable and reproducible method for the assessment of BM hemodilution, several flow cytometry (FC) strategies for hemodilution evaluation have been compared. Methods For each BM sample, cells populations with a well-known distribution in BM and peripheral blood - e.g., mast cells (MC), immature (IG) and mature granulocytes (N) – have been studied by FC and quantified alongside the BM differential count. Results The frequencies of cells’ populations were correlated to the IG/N ratio, highlighting a mild correlation with MCs and erythroblasts (R=0.25 and R=0.38 respectively, with p-value=0.0006 and 0.0000052), whereas no significant correlation was found with B or T-cells. The mild correlation between IG/N, erythroblasts and MCs supported the combined use of these parameters to evaluate BM hemodilution, hence the optimization of the ALLgorithMM. Once validated, the ALLgorithMM was employed to evaluate the dilution status of BM samples in the context of MRD assessment. Overall, we found that 32% of FC and 52% of Next Generation Sequencing (NGS) analyses were MRD negative in samples resulted hemodiluted (HD) or at least mildly hemodiluted (mHD). Conclusions The high frequency of MRD-negative results in both HD and mHD samples implies the presence of possible false negative MRD measurements, impairing the correct assessment of patients’ response to therapy and highlighs the importance to evaluate BM hemodilution.
Collapse
Affiliation(s)
- Ilaria Vigliotta
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
- *Correspondence: Ilaria Vigliotta, ; Carolina Terragna,
| | - Silvia Armuzzi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Martina Barone
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Vincenza Solli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Ignazia Pistis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Enrica Borsi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Barbara Taurisano
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Gaia Mazzocchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Marina Martello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Andrea Poletti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Chiara Sartor
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Ilaria Rizzello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Lucia Pantani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Cristina Papayannidis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
| | - Katia Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Serena Rocchi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Mario Arpinati
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine - University of Bologna, Bologna, Italy
| | - Carolina Terragna
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Seràgnoli Institute of Hematology, Bologna, Italy
- *Correspondence: Ilaria Vigliotta, ; Carolina Terragna,
| |
Collapse
|
15
|
Deng X, Zhang M, Zhou J, Xiao M. Next-generation sequencing for MRD monitoring in B-lineage malignancies: from bench to bedside. Exp Hematol Oncol 2022; 11:50. [PMID: 36057673 PMCID: PMC9440501 DOI: 10.1186/s40164-022-00300-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/21/2022] [Indexed: 12/02/2022] Open
Abstract
Minimal residual disease (MRD) is considered the strongest relevant predictor of prognosis and an effective decision-making factor during the treatment of hematological malignancies. Remarkable breakthroughs brought about by new strategies, such as epigenetic therapy and chimeric antigen receptor-T (CAR-T) therapy, have led to considerably deeper responses in patients than ever, which presents difficulties with the widely applied gold-standard techniques of MRD monitoring. Urgent demands for novel approaches that are ultrasensitive and provide sufficient information have put a spotlight on high-throughput technologies. Recently, advances in methodology, represented by next-generation sequencing (NGS)-based clonality assays, have proven robust and suggestive in numerous high-quality studies and have been recommended by some international expert groups as disease-monitoring modalities. This review demonstrates the applicability of NGS-based clonality assessment for MRD monitoring of B-cell malignancies by summarizing the oncogenesis of neoplasms and the corresponding status of immunoglobulin (IG) rearrangements. Furthermore, we focused on the performance of NGS-based assays compared with conventional approaches and the interpretation of results, revealing directions for improvement and prospects in clinical practice.
Collapse
Affiliation(s)
- Xinyue Deng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Meilan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China.
| |
Collapse
|
16
|
Cho H, Shin S, Chung H, Jang JE, Kim YR, Cheong JW, Min YH, Lee ST, Choi JR, Kim JS. Real-world data on prognostic value of measurable residual disease assessment by fragment analysis or next-generation sequencing in multiple myeloma. Br J Haematol 2022; 198:503-514. [PMID: 35505579 DOI: 10.1111/bjh.18211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 01/12/2023]
Abstract
Measurable residual disease (MRD) negativity is a strong prognostic indicator in multiple myeloma (MM). However, the optimal use of MRD in daily clinical practice has been hampered by the limited feasibility of MRD testing. Therefore, we examined the clinical relevance of commercially available MRD modalities based on clonality assays by fragment analysis with IdentiClone® (n = 73 patients) and next-generation sequencing (NGS) with LymphoTrack® (n = 116 patients) in newly diagnosed patients with MM who received autologous stem cell transplantation (ASCT). MRD was assessed at the end of induction (pre-ASCT) and/or at 100 days after ASCT (post-ASCT). MRD could not predict survival when assessed by fragment analysis. However, NGS-based MRD negativity at pre- or post-ASCT was beneficial in terms of progression-free and overall survival. Moreover, NGS-based MRD negativity was independently associated with improved progression-free and overall survival, and MRD-positive patients both pre- and post-ASCT had worst outcome. Indeed, initial adverse prognostic features by high-risk cytogenetics could be mitigated upon achieving MRD negativity by NGS. We demonstrate the feasibility and clinical benefit of achieving MRD negativity by commercially available clonality-based MRD assays in MM and support incorporating NGS, but not fragment analysis, to tailor therapeutic strategies in real-world practice.
Collapse
Affiliation(s)
- Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Jang
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo Hong Min
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
17
|
Alonso R, Lahuerta JJ. Tumor Reduction in Multiple Myeloma: New Concepts for New Therapeutics. Front Oncol 2022; 11:800309. [PMID: 35096603 PMCID: PMC8794792 DOI: 10.3389/fonc.2021.800309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/23/2021] [Indexed: 12/22/2022] Open
Abstract
The development of new resources for a more accurate diagnosis and response assessment in multiple myeloma has been a long process for decades, mainly since the middle of the 20th century. During this time, the succession of technical advances has run parallel to the better knowledge of disease biology and the availability of novel therapeutic strategies. The cornerstone of standardized criteria to uniformly evaluate the disease response in myeloma dates back to the 1990s when the key role of complete remission was established. Since then, different updates have been implemented according to available scientific evidences not always without certain controversies. The progressive improvements in survival results of myeloma patients and the growing quality of responses due to the novel therapies have led to the need of developing new tools for better monitoring of tumor burden. In this way, the concept of minimal residual disease and its key value based on the prognostic significance and the clinical relevance has been consolidated during the last years, overcoming the value of conventional response criteria or classical adverse prognosis markers. Nevertheless, its precise role in the clinical management of myeloma patients to detect early treatment failure and trigger early rescue strategies is still pending to be defined. In this review, we revisit the major milestones in the understanding of tumor reduction in multiple myeloma until the most recent imaging techniques or liquid biopsy approaches, including a critical view of conventional response criteria, whose backbone has remained unchanged during the last 20 years.
Collapse
Affiliation(s)
- Rafael Alonso
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC CB16/12/00369, Madrid, Spain
| | - Juan José Lahuerta
- Instituto de Investigación Sanitaria, Hospital Universitario 12 de Octubre (imas12) CIBERONC CB16/12/00369, Madrid, Spain
| |
Collapse
|
18
|
Sustained minimal residual disease in myeloma. Blood 2022; 139:469-471. [PMID: 35084473 DOI: 10.1182/blood.2021013199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/13/2021] [Indexed: 11/20/2022] Open
|
19
|
The Exosomes Containing LINC00461 Originated from Multiple Myeloma Inhibit the Osteoblast Differentiation of Bone Mesenchymal Stem Cells via Sponging miR-324-3p. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:3282860. [PMID: 35126917 PMCID: PMC8808147 DOI: 10.1155/2022/3282860] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/24/2021] [Accepted: 12/31/2021] [Indexed: 02/07/2023]
Abstract
Multiple myeloma is one of the hematological malignancies and inhibited osteoblast differentiation of bone marrow mesenchymal stem cells (BM-MSCs) which has been proved as a major complication of the patients with multiple myeloma. However, the pathomechanism of symptom remains unclear. Besides, several studies have indicated that LINC00461 plays an important role in the progression of multiple tumors. Hence, this study attempted to reveal the role of LINC00461 in the osteoblast differentiation of MSCs. In this study, the expression level of LINC00461 in the exosomes of multiple myeloma cells was measured, and BM-MSCs were cultured with the exosomes to observe the change of cellular phenotype. Moreover, downstream target of LINC00461 was searched and verified with dual-luciferase reporter assay, and the activation of the Wnt/β-catenin pathway was also observed by Western blot. The results showed that the isolated BMSCs exhibited special biomarkers of MSCs. LINC00461 was significantly upregulated in the exosomes originated multiple myeloma cells, and increased LINC00461 significantly impeded the osteoblast differentiation of MSCs. Moreover, LINC00461 could significantly suppress the activation of the Wnt/β-catenin pathway in MSCs. In conclusion, this study suggested that LINC00461 in exosomes of multiple myeloma could reduce the activity of the Wnt/β-catenin pathway to inhibit the osteoblast differentiation of BM-MSCs via targeting miR-324-3p.
Collapse
|
20
|
Fitzpatrick MJ, Nardi V, Sohani AR. Plasma cell myeloma: role of histopathology, immunophenotyping, and genetic testing. Skeletal Radiol 2022; 51:17-30. [PMID: 33687521 DOI: 10.1007/s00256-021-03754-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 02/02/2023]
Abstract
Myeloma is a malignant neoplasm of plasma cells with complex pathogenesis. Diagnosis and risk stratification require the integration of histology, radiology, serology, and genetic data. Bone marrow biopsies are essential for myeloma diagnosis by providing material for histologic and cytologic assessment as well as immunophenotypic and genetic studies. Flow cytometry and genetic studies are, in particular, becoming increasingly important for diagnosis, risk stratification, and assessment of treatment response. Myeloma has traditionally been characterized by recurrent cytogenetic abnormalities that can be divided into two subtypes: hyperdiploid, characterized by trisomies, and non-hyperdiploid, characterized by translocations involving chromosome 14. These abnormalities are thought to be primary events, initiating a premalignant state, which progresses to myeloma through the acquisition of secondary mutations. The emergence of next-generation sequencing has led to the discovery of numerous mutations and gene fusions that comprise the heterogenous genomic landscape of myeloma. As the underlying pathogenesis of myeloma continues to be delineated, possible therapeutic targets have also emerged. Herein, we describe the importance of histology, immunophenotype, and mutational analysis in the assessment of myeloma.
Collapse
Affiliation(s)
- Megan J Fitzpatrick
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WRN 219, Boston, MA, 02114, USA
- Department of Pathology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WRN 219, Boston, MA, 02114, USA
- Department of Pathology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Aliyah R Sohani
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, WRN 219, Boston, MA, 02114, USA.
- Department of Pathology, Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA.
| |
Collapse
|
21
|
Mateos MV. High-risk multiple myeloma: how to treat at diagnosis and relapse? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:30-36. [PMID: 34889431 PMCID: PMC8791132 DOI: 10.1182/hematology.2021000229] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Patients with multiple myeloma have experienced a great improvement in survival over the past century because of the introduction of novel therapeutic strategies. However, a subgroup of patients with poorer outcomes than expected is considered high risk and identified by the presence of patient- and disease-based factors such as frailty, extramedullary disease, cytogenetic abnormalities, or even relapses occurring earlier than expected according to the baseline factors. Although the management of patients with high-risk features is not well established because of the lack of specific trials in this subgroup of patients and because of their underrepresentation in the clinical trials, treatment should be planned on 2 pillars: (1) poor prognosis with the presence of high-risk features can be at least improved or even abrogated by achieving a deep and sustained response over time, and (2) this can most likely be obtained through using the best therapeutic options and in a response-adapted way. Some clinical trials that have been planned or are ongoing include only patients with high-risk features, using the most effective therapies (proteasome inhibitors, immunomodulatory drugs, and anti-CD38 monoclonal antibodies) as well as chimeric antigen receptor T cells and T-cell engagers that will unravel what the best therapeutic approach will be to overcome the poor prognosis of the presence of high-risk features.
Collapse
Affiliation(s)
- María-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cancer (IBMCC-USAL, CSIC), Salamanca, Spain
| |
Collapse
|
22
|
Lebel E, Li X, Paul H, Masih-Khan E, Bhella S, Chen C, Prica A, Reece D, Tiedemann R, Trudel S, Kukreti V. Kinetics of response to first- and second-line therapies in multiple myeloma: Assessment by both M-spikes and light chains. Eur J Haematol 2021; 108:204-211. [PMID: 34767270 DOI: 10.1111/ejh.13726] [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: 07/28/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The prognostic value of kinetics of response to multiple myeloma (MM) therapy is controversial. We aimed to expand the knowledge on this topic by reviewing the kinetics of response to both first- and second-line MM therapy, utilizing a homogeneously treated cohort and analyzing separately both M-spike and light chain (LC) responses for each patient. METHODS We reviewed all patients who received first-line cyclophosphamide, bortezomib and dexamethasone induction followed by autologous transplant with melphalan and lenalidomide maintenance in our center between 2007 and 2019. RESULTS Analyzing 360 patients, we observed no correlation between response kinetics to first- versus second-line therapy at the individual patient level. Time to best response to first-line therapy was not a predictor of outcome; however, longer time to best response was highly predictive of a favorable outcome in the second-line setting, independent of other factors. Patients with IgA-MM cleared their M-spike faster than IgG-MM, probably reflecting different half-lives of these isotypes rather than disease biology, as the clearance of LC in both subtypes was similar. CONCLUSIONS Analyzing both M-spike and LC responses in a homogenously treated cohort, we identified important insights regarding the prognostic value of kinetic patterns. Prospective analysis may shed more light on unsolved questions.
Collapse
Affiliation(s)
- Eyal Lebel
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Xuan Li
- Biostatistics Research Unit, University Health Network, Toronto, Ontario, Canada
| | - Harminder Paul
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Esther Masih-Khan
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Sita Bhella
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Christine Chen
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Anca Prica
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Donna Reece
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Rodger Tiedemann
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Suzanne Trudel
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Vishal Kukreti
- Division of Medical Oncology and Hematology at Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Reference Values to Assess Hemodilution and Warn of Potential False-Negative Minimal Residual Disease Results in Myeloma. Cancers (Basel) 2021; 13:cancers13194924. [PMID: 34638406 PMCID: PMC8508317 DOI: 10.3390/cancers13194924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/12/2021] [Accepted: 09/25/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Whereas, in most patients with multiple myeloma (MM), achieving undetectable MRD anticipates a favorable outcome, some others relapse shortly afterwards. Although one obvious explanation for this inconsistency is the use of nonrepresentative marrow samples due to hemodilution, there is no guidance on how to evaluate this issue. METHODS Since B-cell precursors, mast cells and nucleated red blood cells are normally absent in peripheral blood, we analyzed them in 1404 bone marrow (BM) aspirates obtained in numerous disease settings and in 85 healthy adults (HA). RESULTS First, we confirmed the systematic detection of the three populations in HA, as well as the nonreduced numbers with aging. Pairwise comparisons between HA and MM patients grouped according to age and treatment showed significant variability, suggesting that hemodilution should be preferably evaluated with references obtained from patients treated with identical regimens. Leveraging the MRD results from 118 patients, we showed that a comparison with HA of similar age could also inform on potential hemodilution. CONCLUSIONS Our study supports the routine assessment of BM cellularity to evaluate hemodilution, since reduced BM-specific cell types as compared to reference values (either treatment-specific or from HA if the former are unavailable) could indicate hemodilution and a false-negative MRD result.
Collapse
|
24
|
Martinez-Lopez J, Alonso R, Wong SW, Rios R, Shah N, Ruiz-Heredia Y, Sanchez-Pina JM, Sanchez R, Bahri N, Zamanillo I, Poza M, Buenache N, Encinas C, Juarez L, Miras F, Collado L, Barrio S, Martin T, Cedena MT, Wolf J. Making clinical decisions based on measurable residual disease improves the outcome in multiple myeloma. J Hematol Oncol 2021; 14:126. [PMID: 34404440 PMCID: PMC8369640 DOI: 10.1186/s13045-021-01135-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/04/2021] [Indexed: 01/29/2023] Open
Abstract
The assessment of measurable residual disease (MRD) in bone marrow has proven of prognostic relevance in patients with multiple myeloma (MM). Nevertheless, and unlike other hematologic malignancies, the use of MRD results to make clinical decisions in MM has been underexplored to date. In this retrospective study, we present the results from a multinational and multicenter series of 400 patients with MRD monitoring during front-line therapy with the aim of exploring how clinical decisions made based on those MRD results affected outcomes. As expected, achievement of MRD negativity at any point was associated with improved PFS versus persistent MRD positivity (median PFS 104 vs. 45 months, p < 0.0001). In addition, however, 67 out of 400 patients underwent a clinical decision (treatment discontinuation, intensification or initiation of a new therapy) based on MRD results. Those patients in whom a treatment change was made showed a prolonged PFS in comparison with those 333 patients in which MRD results were not acted upon (respectively, mPFS 104 vs. 62 months, p = 0.005). In patients who achieved MRD negativity during maintenance (n = 186) on at least one occasion, stopping therapy in 24 patients vs. continuing in 162 did not alter PFS (mPFS 120 months vs. 82 months, p = 0.1). Most importantly, however, in patients with a positive MRD during maintenance (n = 214), a clinical decision (either intensification or change of therapy) (n = 43) resulted in better PFS compared to patients in whom no adjustment was made (n = 171) (mPFS NA vs. 39 months, p = 0.02). Interestingly, there were no significant differences when MRD was assessed by flow cytometry or by next-generation sequencing. Herein, we find that MRD is useful in guiding clinical decisions during initial therapy and has a positive impact on PFS in MM patients. This potentially opens a new dimension for the use of MRD in MM, but this role still remains to be confirmed in prospective, randomized clinical trials.
Collapse
Affiliation(s)
- Joaquin Martinez-Lopez
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain. .,Department of Medicine, Division of Hematology-Oncology, University of California San Francisco, San Francisco, USA.
| | - Rafael Alonso
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Sandy W Wong
- Department of Medicine, Division of Hematology-Oncology, University of California San Francisco, San Francisco, USA
| | - Rafael Rios
- Hematology Department, Hospital Virgen de Las Nieves, Granada, Spain
| | - Nina Shah
- Department of Medicine, Division of Hematology-Oncology, University of California San Francisco, San Francisco, USA
| | - Yanira Ruiz-Heredia
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Jose Maria Sanchez-Pina
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Ricardo Sanchez
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Natasha Bahri
- Department of Medicine, Division of Hematology-Oncology, University of California San Francisco, San Francisco, USA
| | - Irene Zamanillo
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Maria Poza
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Natalia Buenache
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Cristina Encinas
- Hematology Department, Hospital General Univesitario Gregorio Marañon, Madrid, Spain
| | - Luis Juarez
- Hematology Department, Hospital General Univesitario Gregorio Marañon, Madrid, Spain
| | - Fatima Miras
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Luis Collado
- Medicine Department, Complutense University, Madrid, Spain
| | - Santiago Barrio
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Thomas Martin
- Department of Medicine, Division of Hematology-Oncology, University of California San Francisco, San Francisco, USA
| | - Maria Teresa Cedena
- Hematology Department, Hospital 12 de Octubre i+12, CNIO, Complutense University, Madrid, Spain
| | - Jeffrey Wolf
- Department of Medicine, Division of Hematology-Oncology, University of California San Francisco, San Francisco, USA
| |
Collapse
|
25
|
Romano A, Santoro M, Conticello C, Siragusa S, DI Raimondo F, Martinelli G, Cerchione C. Post-transplant consolidation based on combination of lenalidomide and proteasome inhibitors in multiple myeloma. Panminerva Med 2020; 63:13-20. [PMID: 32955184 DOI: 10.23736/s0031-0808.20.04141-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy due to uncontrolled proliferation of neoplastic plasma cells in the bone marrow, associated to chromosomal instability and cytogenetic abnormalities, which could have an impact on prognosis. Response to treatment and survival of newly diagnosed myeloma patients is heterogeneous, with median overall survival ranging from two to more than ten years, due to clinical and biological factors. To warrant long-term control of disease, several strategies have been proposed in the last years, including short-term high-dose of treatment, named as consolidation, before maintenance. This review will discuss the role of consolidation in the current myeloma treatment landscape, and further improvements required to optimize tailored front-line therapy.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Surgery, University of Catania, Catania, Italy
| | - Marco Santoro
- Department of Surgery, Stomatology and Experimental Oncology, University of Palermo, Palermo, Italy -
| | - Concetta Conticello
- Unit of Hematology, Rodolico San Marco University Polyclinic, Catania, Italy
| | - Sergio Siragusa
- Unit of Hematology, G. D'Alessandro Department of Health Promotion, Mother and Infant Care, Internal and Specialized Medicine, University of Palermo, Palermo, Italy
| | - Francesco DI Raimondo
- Department of Surgery and Medical Surgery, University of Catania, Catania, Italy.,Unit of Hematology, Rodolico San Marco University Polyclinic, Catania, Italy
| | - Giovanni Martinelli
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| |
Collapse
|