1
|
Shen J, Senes F, Wen X, Monti P, Lin S, Pinna C, Murtas A, Podda L, Muntone G, Tidore G, Arru C, Sanna L, Contini S, Virdis P, Sechi LA, Fozza C. Pomalidomide in patients with multiple myeloma: potential impact on the reconstitution of a functional T-cell immunity. Immunol Res 2024; 72:1470-1478. [PMID: 39316338 PMCID: PMC11618177 DOI: 10.1007/s12026-024-09546-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: 03/10/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Pomalidomide, a third-generation oral immunomodulatory drug, exhibits efficacy in patients with relapsed multiple myeloma or those refractory to bortezomib and lenalidomide (RRMM). METHODS In this clinical context, we employed flow cytometry and CDR3 spectratyping to monitor the dynamics of the T-cell repertoire during Pomalidomide treatment, aiming to investigate its potential to reverse the immunological abnormalities characteristic of RRMM. RESULTS By flow cytometry at baseline we found a significant decrease in CD4 + frequency in MM patients, while CD8 + frequency were significantly higher in patients when compared to controls. Most T cell populations remained stable across all time points, except for CD4 + frequency, which notably decreased from t1 to subsequent assessments. Our investigation revealed as most relevant finding the notable increase in CD4 + expansions and the growing prevalence of patients manifesting these expansions. This pattern is even more evident in patients receiving their treatment until t3 and therefore still responding to treatment with Pomalidomide. We also conducted a comparison of spectratyping data before and after treatment, substantially demonstrating a relatively stable pattern throughout the course of Pomalidomide treatment. CONCLUSIONS These observations imply that Pomalidomide treatment influences the T-cell repertoire, particularly in the CD4 + subpopulation during the later stages of treatment, raising speculation about the potential involvement of these lymphocyte expansions in mechanisms related to antitumor immunity.
Collapse
Affiliation(s)
- Jiaxin Shen
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, 515031, Shantou, P. R. China
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Francesca Senes
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Xiaofen Wen
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, 515031, Shantou, P. R. China
| | - Patrizia Monti
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Shaoze Lin
- Department of Hematology, The First Affiliated Hospital of Shantou University Medical College, 515031, Shantou, P. R. China
| | - Claudia Pinna
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Andrea Murtas
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Luigi Podda
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Giuseppina Muntone
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Gianni Tidore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Claudia Arru
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Luca Sanna
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Salvatore Contini
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | - Patrizia Virdis
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy
| | | | - Claudio Fozza
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy.
| |
Collapse
|
2
|
Martinez-Lopez J, Lopez-Muñoz N, Chari A, Dorado S, Barrio S, Arora S, Kumar A, Chung A, Martin T, Wolf J. Measurable residual disease (MRD) dynamics in multiple myeloma and the influence of clonal diversity analyzed by artificial intelligence. Blood Cancer J 2024; 14:131. [PMID: 39112458 PMCID: PMC11306767 DOI: 10.1038/s41408-024-01102-x] [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: 03/22/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Minimal residual disease (MRD) assessment is a known surrogate marker for survival in multiple myeloma (MM). Here, we present a single institution's experience assessing MRD by NGS of Ig genes and the long-term impact of depth of response as well as clonal diversity on the clinical outcome of a large population of MM patients; 482 MM patients at the University of California, San Francisco (UCSF) diagnosed from 2008 to 2020 were analyzed retrospectively. MRD assessment was performed by NGS. PFS curves were plotted by the Kaplan-Meier method. In the newly diagnosed group, 119 of 304, achieved MRD negativity at the level of 10-6 at least once. These patients had a prolonged PFS versus patients who were persistently MRD positive at different levels (p > 0.0001). In the relapsed disease group, 64 of 178 achieved MRD negativity at 10-6, and PFS was prolonged versus patients who remained MRD positive (p = 0.03). Three categories of MRD dynamics were defined by artificial intelligence: (A) patients with ≥3 consistently MRD negative samples, (B) patients with continuously declining but detectable clones, and (C) patients with either increasing or a stable number of clones. Groups A and B had a more prolonged PFS than group C (p < 10-7). Patients who were MRD positive and had not yet relapsed had a higher clonal diversity than those patients who were MRD positive and had relapsed. MRD dynamics can accurately predict disease evolution and drive clinical decision-making. Clonal Diversity could complement MRD assessment in the prediction of outcomes in MM.
Collapse
Affiliation(s)
- J Martinez-Lopez
- Hematology Department, Hospital 12 de Octubre, Complutense University, CNIO, Madrid, Spain.
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - N Lopez-Muñoz
- Hematology Department, Hospital 12 de Octubre, Complutense University, CNIO, Madrid, Spain
| | - A Chari
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - S Dorado
- Medical Department, Altum Sequencing, Madrid, Spain
| | - S Barrio
- Hematology Department, Hospital 12 de Octubre, Complutense University, CNIO, Madrid, Spain
| | - S Arora
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - A Kumar
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - A Chung
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - T Martin
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - J Wolf
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
3
|
Gantana EJ, Musekwa E, Chapanduka ZC. Advances in estimating plasma cells in bone marrow: A comprehensive method review. Afr J Lab Med 2024; 13:2381. [PMID: 39114749 PMCID: PMC11304106 DOI: 10.4102/ajlm.v13i1.2381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/25/2024] [Indexed: 08/10/2024] Open
Abstract
The quantitation of plasma cells in bone marrow (BM) is crucial for diagnosing and classifying plasma cell neoplasms. Various methods, including Romanowsky-stained BM aspirates (BMA), immunohistochemistry, flow cytometry, and radiological imaging, have been explored. However, challenges such as patchy infiltration and sample haemodilution can impact the reliability of BM plasma cell percentage estimates. Bone marrow plasma cell percentage varies across methods, with immunohistochemically stained biopsies consistently yielding higher values than Romanowsky-stained BMA or flow cytometry alone. CD138 or MUM1 immunohistochemistry and artificial intelligence image analysis on whole-slide images are emerging as promising tools for accurate plasma cell identification and quantification. Radiological imaging, particularly with advanced technologies like dual-energy computed tomography and radiomics, shows potential for multiple myeloma diagnosis, although standardisation remains a challenge. Molecular techniques, such as allele-specific oligonucleotide quantitative polymerase chain reaction and next-generation sequencing, offer insights into clonality and measurable residual disease. While no consensus exists on a gold standard method for BM plasma cell quantitation, CD138-stained biopsies are favoured for accurate estimation and play a pivotal role in diagnosing and assessing multiple myeloma treatment responses. Combining multiple methods, such as BMA, BM biopsy, and flow cytometry, enhances accuracy of diagnosis and classification of plasma cell neoplasms. The quest for a gold standard requires ongoing research and collaboration to refine existing methods. Furthermore, the rise of digital pathology is anticipated to reshape laboratory medicine and the role of pathologists in the digital era. What this study adds This article adds a comprehensive review and comparison of different methods for plasma cell estimation in the bone marrow, highlighting their strengths and limitations. The goal is to contribute valuable insights that can guide the selection of optimal techniques for accurate plasma cell estimation.
Collapse
Affiliation(s)
- Ethan J Gantana
- Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Haematology, National Health Laboratory Service, Cape Town, South Africa
| | - Ernest Musekwa
- Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Haematology, National Health Laboratory Service, Cape Town, South Africa
| | - Zivanai C Chapanduka
- Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Department of Haematology, National Health Laboratory Service, Cape Town, South Africa
| |
Collapse
|
4
|
Medina-Herrera A, Sarasquete ME, Jiménez C, Puig N, García-Sanz R. Minimal Residual Disease in Multiple Myeloma: Past, Present, and Future. Cancers (Basel) 2023; 15:3687. [PMID: 37509348 PMCID: PMC10377959 DOI: 10.3390/cancers15143687] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Responses to treatment have improved over the last decades for patients with multiple myeloma. This is a consequence of the introduction of new drugs that have been successfully combined in different clinical contexts: newly diagnosed, transplant-eligible or ineligible patients, as well as in the relapsed/refractory setting. However, a great proportion of patients continue to relapse, even those achieving complete response, which underlines the need for updated response criteria. In 2014, the international myeloma working group established new levels of response, prompting the evaluation of minimal residual disease (MRD) for those patients already in complete or stringent complete response as defined by conventional serological assessments: the absence of tumor plasma cells in 100,000 total cells or more define molecular and immunophenotypic responses by next-generation sequencing and flow cytometry, respectively. In this review, we describe all the potential methods that may be used for MRD detection based on the evidence found in the literature, paying special attention to their advantages and pitfalls from a critical perspective.
Collapse
Affiliation(s)
- Alejandro Medina-Herrera
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - María Eugenia Sarasquete
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Cristina Jiménez
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Noemí Puig
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Ramón García-Sanz
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| |
Collapse
|
5
|
Marx A, Osváth M, Szikora B, Pipek O, Csabai I, Nagy Á, Bödör C, Matula Z, Nagy G, Bors A, Uher F, Mikala G, Vályi-Nagy I, Kacskovics I. Liquid biopsy-based monitoring of residual disease in multiple myeloma by analysis of the rearranged immunoglobulin genes-A feasibility study. PLoS One 2023; 18:e0285696. [PMID: 37235573 DOI: 10.1371/journal.pone.0285696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
The need for sensitive monitoring of minimal/measurable residual disease (MRD) in multiple myeloma emerged as novel therapies led to deeper responses. Moreover, the potential benefits of blood-based analyses, the so-called liquid biopsy is prompting more and more studies to assess its feasibility. Considering these recent demands, we aimed to optimize a highly sensitive molecular system based on the rearranged immunoglobulin (Ig) genes to monitor MRD from peripheral blood. We analyzed a small group of myeloma patients with the high-risk t(4;14) translocation, using next-generation sequencing of Ig genes and droplet digital PCR of patient-specific Ig heavy chain (IgH) sequences. Moreover, well established monitoring methods such as multiparametric flow cytometry and RT-qPCR of the fusion transcript IgH::MMSET (IgH and multiple myeloma SET domain-containing protein) were utilized to evaluate the feasibility of these novel molecular tools. Serum measurements of M-protein and free light chains together with the clinical assessment by the treating physician served as routine clinical data. We found significant correlation between our molecular data and clinical parameters, using Spearman correlations. While the comparisons of the Ig-based methods and the other monitoring methods (flow cytometry, qPCR) were not statistically evaluable, we found common trends in their target detection. Regarding longitudinal disease monitoring, the applied methods yielded complementary information thus increasing the reliability of MRD evaluation. We also detected indications of early relapse before clinical signs, although this implication needs further verification in a larger patient cohort.
Collapse
Affiliation(s)
- Anita Marx
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Magdolna Osváth
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Bence Szikora
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - István Csabai
- Department of Physics of Complex Systems, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Ákos Nagy
- Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- Department of Pathology and Experimental Cancer Research, HCEMM-SE Molecular Oncohematology Research Group, Semmelweis University, Budapest, Hungary
| | - Zsolt Matula
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Ginette Nagy
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - András Bors
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Ferenc Uher
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Gábor Mikala
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - István Vályi-Nagy
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, Budapest, Hungary
| | - Imre Kacskovics
- Department of Immunology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
6
|
Nau A, Shen Y, Sanchorawala V, Prokaeva T, Morgan GJ. Complete variable domain sequences of monoclonal antibody light chains identified from untargeted RNA sequencing data. Front Immunol 2023; 14:1167235. [PMID: 37143670 PMCID: PMC10151772 DOI: 10.3389/fimmu.2023.1167235] [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: 02/16/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Introduction Monoclonal antibody light chain proteins secreted by clonal plasma cells cause tissue damage due to amyloid deposition and other mechanisms. The unique protein sequence associated with each case contributes to the diversity of clinical features observed in patients. Extensive work has characterized many light chains associated with multiple myeloma, light chain amyloidosis and other disorders, which we have collected in the publicly accessible database, AL-Base. However, light chain sequence diversity makes it difficult to determine the contribution of specific amino acid changes to pathology. Sequences of light chains associated with multiple myeloma provide a useful comparison to study mechanisms of light chain aggregation, but relatively few monoclonal sequences have been determined. Therefore, we sought to identify complete light chain sequences from existing high throughput sequencing data. Methods We developed a computational approach using the MiXCR suite of tools to extract complete rearranged IGVL-IGJL sequences from untargeted RNA sequencing data. This method was applied to whole-transcriptome RNA sequencing data from 766 newly diagnosed patients in the Multiple Myeloma Research Foundation CoMMpass study. Results Monoclonal IGVL-IGJL sequences were defined as those where >50% of assigned IGK or IGL reads from each sample mapped to a unique sequence. Clonal light chain sequences were identified in 705/766 samples from the CoMMpass study. Of these, 685 sequences covered the complete IGVL-IGJL region. The identity of the assigned sequences is consistent with their associated clinical data and with partial sequences previously determined from the same cohort of samples. Sequences have been deposited in AL-Base. Discussion Our method allows routine identification of clonal antibody sequences from RNA sequencing data collected for gene expression studies. The sequences identified represent, to our knowledge, the largest collection of multiple myeloma-associated light chains reported to date. This work substantially increases the number of monoclonal light chains known to be associated with non-amyloid plasma cell disorders and will facilitate studies of light chain pathology.
Collapse
Affiliation(s)
- Allison Nau
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Yun Shen
- Research Computing Services, Boston University, Boston, MA, United States
| | - Vaishali Sanchorawala
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Tatiana Prokaeva
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Gareth J. Morgan
- Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Section of Hematology and Medical Oncology, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
7
|
Single-Cell RNA Sequencing for the Detection of Clonotypic V(D)J Rearrangements in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232415691. [PMID: 36555330 PMCID: PMC9779610 DOI: 10.3390/ijms232415691] [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: 11/15/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) has a highly heterogeneous genetic background, which complicates its molecular tracking over time. Nevertheless, each MM patient's malignant plasma cells (PCs) share unique V(D)J rearranged sequences at immunoglobulin loci, which represent ideal disease biomarkers. Because the tumor-specific V(D)J sequence is highly expressed in bulk RNA in MM patients, we wondered whether it can be identified by single-cell RNA sequencing (scRNA-seq). To this end we analyzed CD138+ cells purified from bone marrow aspirates of 19 samples with PC dyscrasias by both a standard method based on bulk DNA and by an implementation of the standard 10x Genomics protocol to detect expressed V(D)J sequences. A dominant clonotype was easily identified in each sample, accounting on average for 83.65% of V(D)J-rearranged cells. Compared with standard methods, scRNA-seq analysis proved highly concordant and even more effective in identifying clonal productive rearrangements, by-passing limitations related to the misannealing of consensus primers in hypermutated regions. We next validated its accuracy to track 5 clonal cells with absolute sensitivity in a virtual sample containing 3180 polyclonal cells. This shows that single-cell V(D)J analysis may be used to find rare clonal cells, laying the foundations for functional single-cell dissection of minimal residual disease.
Collapse
|
8
|
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: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [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
|
9
|
Hultcrantz M, Rustad EH, Yellapantula V, Jacob A, Akhlaghi T, Korde N, Mailankody S, Lesokhin AM, Hassoun H, Smith EL, Lahoud OB, Landau HJ, Shah GL, Scordo M, Chung DJ, Giralt S, Papaemmanuil E, Landgren O. Capture Rate of V(D)J Sequencing for Minimal Residual Disease Detection in Multiple Myeloma. Clin Cancer Res 2022; 28:2160-2166. [PMID: 35553646 DOI: 10.1158/1078-0432.ccr-20-2995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/28/2020] [Accepted: 02/18/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Minimal residual disease (MRD) negativity is a strong predictor for outcome in multiple myeloma. To assess V(D)J clonotype capture using the updated Adaptive next-generation sequencing (NGS) MRD assay in a clinical setting, we analyzed baseline and follow-up samples from patients with multiple myeloma who achieved deep clinical responses. EXPERIMENTAL DESIGN A total of 159 baseline and 31 follow-up samples from patients with multiple myeloma were sequenced using the NGS MRD assay. Baseline samples were also sequenced using a targeted multiple myeloma panel (myTYPE). We estimated ORs with 95% confidence intervals (CI) for clonotypes detection using logistic regression. RESULTS The V(D)J clonotype capture rate was 93% in baseline samples with detectable genomic aberrations, indicating presence of tumor DNA, assessed through myTYPE. myTYPE-positive samples had significantly higher V(D)J clonotype detection rates in univariate (OR, 7.3; 95% CI, 2.8-22.6) and multivariate analysis (OR, 4.4; 95% CI, 1.4-16.9; P = 0.016). Higher disease burden was associated with higher probability of V(D)J clonotype capture, meanwhile no such association was found for age, gender, or type of heavy or light immunoglobulin chain. All V(D)J clonotypes detected at baseline were detected in MRD-positive samples indicating that the V(D)J clonotypes remained stable and did not undergo further rearrangements during follow-up. Of the 31 posttreatment samples, 12 were MRD-negative using the NGS MRD assay. CONCLUSIONS NGS for V(D)J rearrangements in multiple myeloma offers a reliable and sensitive method for MRD tracking with high detection rates in the clinical setting.
Collapse
Affiliation(s)
- Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York.,Karolinska Institute, Department of Medicine, Solna, Stockholm, Sweden
| | - Even H Rustad
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Venkata Yellapantula
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | | | - Theresia Akhlaghi
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Alexander M Lesokhin
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Hani Hassoun
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Eric L Smith
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Oscar B Lahoud
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Heather J Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - David J Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Sergio Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Manhattan, New York
| | - Elli Papaemmanuil
- Department of Cancer Immunology, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
10
|
He H, Li Z, Lu J, Qiang W, Jiang S, Xu Y, Fu W, Zhai X, Zhou L, Qian M, Du J. Single-cell RNA-seq reveals clonal diversity and prognostic genes of relapsed multiple myeloma. Clin Transl Med 2022; 12:e757. [PMID: 35297204 PMCID: PMC8926895 DOI: 10.1002/ctm2.757] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a clinically and biologically heterogeneous plasma-cell malignancy. Despite extensive research, disease heterogeneity and relapse remain a big challenge in MM therapeutics. We tried to dissect this disease and identify novel biomarkers for patient stratification and treatment outcome prediction by applying single-cell technology. METHODS We performed single-cell RNA sequencing (scRNA-seq) and variable-diversity-joining regions-targeted sequencing (scVDJ-seq) concurrently on bone marrow samples from a cohort of 18 patients with newly diagnosed MM (NDMM; n = 12) or refractory/relapsed MM (RRMM; n = 6). We analysed the malignant clonotypes using scVDJ-seq data and conducted data integration and cell-type annotation through the CCA algorithm based on gene expression profiling. Furthermore, we identified disease status-specific genes and modules by comparison of NDMM and RRMM datasets and explored the findings in a larger MM cohort from the MMRF CoMMpass study. RESULTS We found that all the myeloma cells in either diagnosed or relapsed samples were dominated by a major clone, with a few subclones in several samples (n = 5). Next, we investigated the universal transcriptional features of myeloma cells and identified eight meta-programs correlated with this disease, especially meta-programs 1 and 8 (M1 and M8), which were the most significant and related to cell cycle and stress response, respectively. Furthermore, we classified the malignant plasma cells into eight clusters and found that the cell numbers in clusters 2/6/7 were exclusively higher in relapsed samples. Besides, we identified several attractive candidates for biomarkers (e.g. SMAD1 and STMN1) associated with disease progression and relapse in our dataset and related to overall survival in the CoMMpass dataset. CONCLUSIONS Our data provide insights into the heterogeneity of MM as well as highlight the relevance of intra-tumour heterogeneity and discover novel biomarkers that might be a potent therapy.
Collapse
Affiliation(s)
- Haiyan He
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Zifeng Li
- Institute of Pediatrics and Department of Hematology and OncologyChildren's Hospital of Fudan UniversityNational Children's Medical CenterShanghaiChina
| | - Jing Lu
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Wanting Qiang
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Sihan Jiang
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Yaochen Xu
- Shanghai Key Laboratory of Medical Epigenetics, International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Weijun Fu
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Xiaowen Zhai
- Institute of Pediatrics and Department of Hematology and OncologyChildren's Hospital of Fudan UniversityNational Children's Medical CenterShanghaiChina
| | - Lin Zhou
- Department of Laboratory MedicineChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and OncologyChildren's Hospital of Fudan UniversityNational Children's Medical CenterShanghaiChina
| | - Juan Du
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| |
Collapse
|
11
|
Ruan GX, Li Y, Chen W, Huang H, Zhang R, Chen C, Lam KP, Xu S, Ou X. The spliceosome component Usp39 controls B cell development by regulating immunoglobulin gene rearrangement. Cell Rep 2022; 38:110338. [PMID: 35139388 DOI: 10.1016/j.celrep.2022.110338] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 11/18/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
The spliceosome is a large ribonucleoprotein complex responsible for pre-mRNA splicing and genome stability maintenance. Disruption of the spliceosome activity may lead to developmental disorders and tumorigenesis. However, the physiological role that the spliceosome plays in B cell development and function is still poorly defined. Here, we demonstrate that ubiquitin-specific peptidase 39 (Usp39), a spliceosome component of the U4/U6.U5 tri-snRNP complex, is essential for B cell development. Ablation of Usp39 in B cell lineage blocks pre-pro-B to pro-B cell transition in the bone marrow, leading to a profound reduction of mature B cells in the periphery. We show that Usp39 specifically regulates immunoglobulin gene rearrangement in a spliceosome-dependent manner, which involves modulating chromatin interactions at the Igh locus. Moreover, our results indicate that Usp39 deletion reduces the pre-malignant B cells in Eμ-Myc transgenic mice and significantly improves their survival.
Collapse
Affiliation(s)
- Gui-Xin Ruan
- Harbin Institute of Technology, Harbin 150001, China; Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuxing Li
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wenjing Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hengjun Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Rui Zhang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Changxu Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; Departments of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xijun Ou
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
12
|
Mack EKM, Hartmann S, Ross P, Wollmer E, Mann C, Neubauer A, Brendel C, Hoffmann J. Monitoring multiple myeloma in the peripheral blood based on cell-free DNA and circulating plasma cells. Ann Hematol 2022; 101:811-824. [PMID: 35106639 PMCID: PMC8913458 DOI: 10.1007/s00277-022-04771-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/16/2022] [Indexed: 11/25/2022]
Abstract
With the advent of novel, highly effective therapies for multiple myeloma (MM), classical serologic monitoring appears insufficient for response assessment and prediction of relapse. Moreover, serologic studies in MM are hampered by interference of therapeutic antibodies. The detection of malignant plasma cell clones by next generation sequencing (NGS) or multiparameter flow cytometry (MFC) circumvents these difficulties and can be performed in the peripheral blood (pB) by targeting circulating cell-free DNA (cfDNA) or circulating plasma cells (CPCs), thus also avoiding an invasive sampling procedure. Here, we applied NGS of VJ light chain (LC) rearrangements in cfDNA and MFC of magnetically-enriched CD138-positive CPCs (me-MFC) to investigate disease burden in unselected MM patients. Sequencing was successful for 114/130 (87.7%) cfDNA samples and me-MFC results were analyzable for 196/205 (95.6%) samples. MM clones were detectable in 38.9% of samples taken at initial diagnosis or relapse (ID/RD), but only in 11.8% of samples taken during complete remission (CR). Circulating MM plasma cells were present in 83.3% of ID/RD samples and 9.9% of CR samples. Residual disease assessment by NGS or me-MFC in samples taken during very good partial remission or CR was 80% concordant. Notably, 4/4 (NGS) and 5/8 (me-MFC) positive CR samples were from patients with oligo- or non-secretory myeloma. The time to progression was shorter if there was evidence of residual myeloma in the pB. Together, our findings indicate that our two novel analytical approaches accurately indicate the course of MM and may be particularly valuable for monitoring patients with serologically non-trackable disease.
Collapse
Affiliation(s)
- Elisabeth K M Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| | - Sören Hartmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Petra Ross
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Ellen Wollmer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Christoph Mann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Jörg Hoffmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| |
Collapse
|
13
|
Charalampous C, Kourelis T. Minimal Residual Disease Assessment in Multiple Myeloma Patients: Minimal Disease With Maximal Implications. Front Oncol 2022; 11:801851. [PMID: 35155198 PMCID: PMC8825476 DOI: 10.3389/fonc.2021.801851] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/29/2021] [Indexed: 11/21/2022] Open
Abstract
Multiple Myeloma (MM), the second most common hematologic malignancy, has been the target of many therapeutic advances over the past two decades. The introduction of novel agents, such as proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies, along with autologous hematopoietic stem cell transplantation (ASCT) in the current standard of care, has increased the median survival of myeloma patients significantly. Nevertheless, a curative treatment option continues to elude us, and MM remains an incurable disease, with patients relapsing even after achieving deep conventionally defined responses, underscoring the need for the development of sensitive methods that will allow for proper identification and management of the patients with a higher probability of relapse. Accurate detection of Minimal Residual Disease (MRD) from a bone marrow biopsy represents a relatively new approach of evaluating response to treatment with data showing clear benefit from obtaining MRD(-) status at any point of the disease course. As life expectancy for patients with MM continues to increase and deep responses are starting to become the norm, establishing and refining the role of MRD in the disease course is more relevant than ever. This review examines the different methods used to detect MRD and discusses future considerations regarding the implementation in day-to-day clinical practice and as a prospective primary endpoint for clinical trials.
Collapse
|
14
|
Cai G, Guan Z, Jin Y, Su Z, Chen X, Liu Q, Wang C, Yin X, Zhang L, Ye G, Luo W. Circulating T-Cell Repertoires Correlate With the Tumor Response in Patients With Breast Cancer Receiving Neoadjuvant Chemotherapy. JCO Precis Oncol 2022; 6:e2100120. [PMID: 35025620 PMCID: PMC8769146 DOI: 10.1200/po.21.00120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Neoadjuvant chemotherapy (NAC) has been widely used in patients with breast cancer to minish tumor burden and increase resection rate of cancer. T-cell repertoire has been believed to be able to monitor antitumor immune responses. This study aimed to explore the dynamic change of T-cell repertoire and its clinical value in evaluating the tumor response in patients with breast cancer receiving NAC. MATERIALS AND METHODS Ninety-four patients who underwent NAC before surgery were recruited, and peripheral blood samples were collected at multiple time points during NAC. High-throughput T-cell receptor (TCR)-β sequencing was used to characterize the T-cell repertoire of every sample and analyzed the changes in circulating T-cell repertoire during NAC. RESULTS We found that the diversity of TCR repertoires was associated with age and clinical stage of the patients with breast cancer. The distribution of Vβ and Jβ genes in TCR repertoires was skewed in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer. Vβ20.1 and Vβ30 expression levels before NAC correlate with tumor response after all cycles of NAC in HER2- and HER2+ patients, respectively. Some CDR3 motifs that correlated with clinical response in either HER2+ or HER2- patients were identified. Besides, TCR repertoire evolved during NAC and the diversity of TCR repertoire decreased more after two cycles of NAC in patients with good tumor response after all cycles of NAC (P = .0061). CONCLUSION Our results demonstrated that TCR repertoire correlated with the characteristics of the tumor, such as the expression status of HER2. Moreover, some characteristics of TCR repertoires that correlated with clinical response were identified and they might provide useful information to tailor therapeutic regimens at the early cycle of NAC.
Collapse
Affiliation(s)
- Gengxi Cai
- The First People's Hospital of Foshan, Foshan, China
| | - Zhanwen Guan
- The First People's Hospital of Foshan, Foshan, China
| | - Yabin Jin
- The First People's Hospital of Foshan, Foshan, China
| | - Zuhui Su
- The First People's Hospital of Foshan, Foshan, China
| | | | - Qing Liu
- The First People's Hospital of Foshan, Foshan, China
| | | | - Xiaoxia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, China
| | - Lifang Zhang
- The First People's Hospital of Foshan, Foshan, China
| | - Guolin Ye
- The First People's Hospital of Foshan, Foshan, China
| | - Wei Luo
- The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
15
|
Clinical value of measurable residual disease testing for assessing depth, duration, and direction of response in multiple myeloma. Blood Adv 2021; 4:3295-3301. [PMID: 32706892 DOI: 10.1182/bloodadvances.2020002037] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/12/2020] [Indexed: 12/21/2022] Open
Abstract
Few clinical studies have reported results of measurable residual disease (MRD) assessments performed as part of routine practice. Herein we present our single-institution experience assessing MRD in 234 multiple myeloma (MM) patients (newly diagnosed [NDMM = 159] and relapsed [RRMM = 75]). We describe the impact of depth, duration, and direction of response on prognosis. MRD assessments were performed by next-generation sequencing of immunoglobulin genes with a sensitivity of 10-6. Those achieving MRD negativity at 10-6, as well as 10-5, had superior median progression-free survival (PFS). In the NDMM cohort, 40% of the patients achieved MRD negativity at 10-6 and 59% at 10-5. Median PFS in the NDMM cohort was superior in those achieving MRD at 10-5 vs <10-5 (PFS: 87 months vs 32 months; P < .001). In the RRMM cohort, 36% achieved MRD negativity at 10-6 and 47% at 10-5. Median PFS was superior for the RRMM achieving MRD at 10-5 vs <10-5 (PFS: 42 months vs 17 months; P < .01). Serial MRD monitoring identified 3 categories of NDMM patients: (A) patients with ≥3 MRD 10-6 negative samples, (B) patients with detectable but continuously declining clonal numbers, and (C) patients with stable or increasing clonal number (≥1 log). PFS was superior in groups A and B vs C (median PFS not reached [NR], NR, 55 respectively; P < .001). This retrospective evaluation of MRD used as part of clinical care validates MRD as an important prognostic marker in NDMM and RRMM and supports its use as an endpoint in future clinical trials as well as for clinical decision making.
Collapse
|
16
|
Maclachlan KH, Came N, Diamond B, Roshal M, Ho C, Thoren K, Mayerhoefer ME, Landgren O, Harrison S. Minimal residual disease in multiple myeloma: defining the role of next generation sequencing and flow cytometry in routine diagnostic use. Pathology 2021; 53:385-399. [PMID: 33674146 DOI: 10.1016/j.pathol.2021.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
For patients diagnosed with multiple myeloma (MM) there have been significant treatment advances over the past decade, reflected in an increasing proportion of patients achieving durable remissions. Clinical trials repeatedly demonstrate that achieving a deep response to therapy, with a bone marrow assessment proving negative for minimal residual disease (MRD), confers a significant survival advantage. To accurately assess for minute quantities of residual cancer requires highly sensitive methods; either multiparameter flow cytometry or next generation sequencing are currently recommended for MM response assessment. Under optimal conditions, these methods can detect one aberrant cell amongst 1,000,000 normal cells (a sensitivity of 10-6). Here, we will review the practical use of MRD assays in MM, including challenges in implementation for the routine diagnostic laboratory, standardisation across laboratories and clinical trials, the clinical integration of MRD status assessment into MM management and future directions for ongoing research.
Collapse
Affiliation(s)
- Kylee H Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Haematology Service, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia.
| | - Neil Came
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia; Pathology Department, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia
| | - Benjamin Diamond
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katie Thoren
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Myeloma Program, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Simon Harrison
- Haematology Service, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
17
|
Diamond BT, Rustad E, Maclachlan K, Thoren K, Ho C, Roshal M, Ulaner GA, Landgren CO. Defining the undetectable: The current landscape of minimal residual disease assessment in multiple myeloma and goals for future clarity. Blood Rev 2021; 46:100732. [PMID: 32771227 PMCID: PMC9928431 DOI: 10.1016/j.blre.2020.100732] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/05/2020] [Accepted: 07/06/2020] [Indexed: 01/19/2023]
Abstract
Multiple Myeloma, the second most prevalent hematologic malignancy, yet lacks an established curative therapy. However, overall response rate to modern four-drug regimens approaches 100%. Major efforts have thus focused on the measurement of minute quantities of residual disease (minimal residual disease or MRD) for prognostic metrics and therapeutic response evaluation. Currently, MRD is assessed by flow cytometry or by next generation sequencing to track tumor-specific immunoglobulin V(D)J rearrangements. These bone marrow-based methods can reach sensitivity thresholds of the identification of one neoplastic cell in 1,000,000 (10-6). New technologies are being developed to be used alone or in conjunction with established methods, including peripheral blood-based assays, mass spectrometry, and targeted imaging. Data is also building for MRD as a surrogate endpoint for overall survival. Here, we will address the currently utilized MRD assays, challenges in validation across labs and clinical trials, techniques in development, and future directions for successful clinical application of MRD in multiple myeloma.
Collapse
Affiliation(s)
| | | | | | | | - Caleb Ho
- Memorial Sloan Kettering Cancer Center, USA
| | | | | | | |
Collapse
|
18
|
Zuo X, Liu D. Progress in the application of minimal residual disease detection in multiple myeloma. J Hematop 2021. [DOI: 10.1007/s12308-020-00436-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
19
|
Medina-Martínez JS, Arango-Ossa JE, Levine MF, Zhou Y, Gundem G, Kung AL, Papaemmanuil E. Isabl Platform, a digital biobank for processing multimodal patient data. BMC Bioinformatics 2020; 21:549. [PMID: 33256603 PMCID: PMC7708092 DOI: 10.1186/s12859-020-03879-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The widespread adoption of high throughput technologies has democratized data generation. However, data processing in accordance with best practices remains challenging and the data capital often becomes siloed. This presents an opportunity to consolidate data assets into digital biobanks-ecosystems of readily accessible, structured, and annotated datasets that can be dynamically queried and analysed. RESULTS We present Isabl, a customizable plug-and-play platform for the processing of multimodal patient-centric data. Isabl's architecture consists of a relational database (Isabl DB), a command line client (Isabl CLI), a RESTful API (Isabl API) and a frontend web application (Isabl Web). Isabl supports automated deployment of user-validated pipelines across the entire data capital. A full audit trail is maintained to secure data provenance, governance and ensuring reproducibility of findings. CONCLUSIONS As a digital biobank, Isabl supports continuous data utilization and automated meta analyses at scale, and serves as a catalyst for research innovation, new discoveries, and clinical translation.
Collapse
Affiliation(s)
| | | | - Max F Levine
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yangyu Zhou
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gunes Gundem
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew L Kung
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
20
|
Baseline VDJ clonotype detection using a targeted sequencing NGS assay: allowing for subsequent MRD assessment. Blood Cancer J 2020; 10:76. [PMID: 32699232 PMCID: PMC7376205 DOI: 10.1038/s41408-020-00343-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/24/2020] [Accepted: 04/30/2020] [Indexed: 12/27/2022] Open
|
21
|
Rustad EH, Yellapantula V, Leongamornlert D, Bolli N, Ledergor G, Nadeu F, Angelopoulos N, Dawson KJ, Mitchell TJ, Osborne RJ, Ziccheddu B, Carniti C, Montefusco V, Corradini P, Anderson KC, Moreau P, Papaemmanuil E, Alexandrov LB, Puente XS, Campo E, Siebert R, Avet-Loiseau H, Landgren O, Munshi N, Campbell PJ, Maura F. Timing the initiation of multiple myeloma. Nat Commun 2020; 11:1917. [PMID: 32317634 PMCID: PMC7174344 DOI: 10.1038/s41467-020-15740-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
The evolution and progression of multiple myeloma and its precursors over time is poorly understood. Here, we investigate the landscape and timing of mutational processes shaping multiple myeloma evolution in a large cohort of 89 whole genomes and 973 exomes. We identify eight processes, including a mutational signature caused by exposure to melphalan. Reconstructing the chronological activity of each mutational signature, we estimate that the initial transformation of a germinal center B-cell usually occurred during the first 2nd-3rd decades of life. We define four main patterns of activation-induced deaminase (AID) and apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC) mutagenesis over time, including a subset of patients with evidence of prolonged AID activity during the pre-malignant phase, indicating antigen-responsiveness and germinal center reentry. Our findings provide a framework to study the etiology of multiple myeloma and explore strategies for prevention and early detection.
Collapse
Affiliation(s)
- Even H Rustad
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Venkata Yellapantula
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Leongamornlert
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Niccolò Bolli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Guy Ledergor
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Ferran Nadeu
- Patologia Molecular de Neoplàsies Limfoides, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
| | - Nicos Angelopoulos
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Kevin J Dawson
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Thomas J Mitchell
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Robert J Osborne
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Bachisio Ziccheddu
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Turin, Turin, Italy
| | - Cristiana Carniti
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Vittorio Montefusco
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Corradini
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Philippe Moreau
- CRCINA, SIRIC ILIAD, University Hospital of Nantes, Nantes, France
| | - Elli Papaemmanuil
- Computational Oncology Service, Department of Epidemiology & Biostatistics, Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California, La Jolla, San Diego, CA, USA
| | - Xose S Puente
- Unitat Hematopatologia, Hospital Clínic of Barcelona, Universitat de Barcelona, 08036, Barcelona, Spain
- Departamento de Bioquimica y Biologia Molecular, Instituto Universitario de Oncologia (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Elias Campo
- Patologia Molecular de Neoplàsies Limfoides, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029, Madrid, Spain
- Unitat Hematopatologia, Hospital Clínic of Barcelona, Universitat de Barcelona, 08036, Barcelona, Spain
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | | | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Veterans Administration Boston Healthcare System, West Roxbury, MA, USA
| | - Peter J Campbell
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Francesco Maura
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- The Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK.
| |
Collapse
|
22
|
Rustad EH, Boyle EM. Monitoring minimal residual disease in the bone marrow using next generation sequencing. Best Pract Res Clin Haematol 2020; 33:101149. [PMID: 32139014 DOI: 10.1016/j.beha.2020.101149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/25/2022]
Abstract
Achieving minimal residual disease (MRD) negativity in the bone marrow is one of the strongest prognostic factors in multiple myeloma. Consequently, MRD testing is routinely performed in clinical trials and moving towards standard of care. This review focuses on the role of next generation sequencing (NGS) of tumor-specific immunoglobulin V(D)J sequences for MRD tracking. The immunoglobulin variable regions are ideal targets for tracking, because every tumor cell shares an identical gene sequence, which is stable over time and generally distinct from the immunoglobulin sequences of normal B-cells. Several excellent assays for NGS-based MRD testing are available, both commercial and community-based, including one that is FDA-approved. These assays can achieve the gold standard analytical sensitivity of one tumor cell per million (10-6), requiring a minimum input of 3 million bone marrow cells. On-going clinical trials will outline how MRD testing should be used to inform dynamic risk-adopted therapy.
Collapse
Affiliation(s)
- Even H Rustad
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eileen M Boyle
- Myeloma Research Program, NYU Langone Perlmutter Cancer Center, NYC, NY, 10016, USA
| |
Collapse
|
23
|
Comprehensive detection of recurring genomic abnormalities: a targeted sequencing approach for multiple myeloma. Blood Cancer J 2019; 9:101. [PMID: 31827071 PMCID: PMC6906304 DOI: 10.1038/s41408-019-0264-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
Recent genomic research efforts in multiple myeloma have revealed clinically relevant molecular subgroups beyond conventional cytogenetic classifications. Implementing these advances in clinical trial design and in routine patient care requires a new generation of molecular diagnostic tools. Here, we present a custom capture next-generation sequencing (NGS) panel designed to identify rearrangements involving the IGH locus, arm level, and focal copy number aberrations, as well as frequently mutated genes in multiple myeloma in a single assay. We sequenced 154 patients with plasma cell disorders and performed a head-to-head comparison with the results from conventional clinical assays, i.e., fluorescent in situ hybridization (FISH) and single-nucleotide polymorphism (SNP) microarray. Our custom capture NGS panel had high sensitivity (>99%) and specificity (>99%) for detection of IGH translocations and relevant chromosomal gains and losses in multiple myeloma. In addition, the assay was able to capture novel genomic markers associated with poor outcome such as bi-allelic events involving TP53. In summary, we show that a multiple myeloma designed custom capture NGS panel can detect IGH translocations and CNAs with very high concordance in relation to FISH and SNP microarrays and importantly captures the most relevant and recurrent somatic mutations in multiple myeloma rendering this approach highly suitable for clinical application in the modern era.
Collapse
|