51
|
Perroud C, Thurian D, Andres M, Künzi A, Wiedemann G, Zeerleder S, Bacher U, Pabst T, Banz Y, Porret N, Rebmann E. Effect of MAPK activation via mutations in NRAS, KRAS and BRAF on clinical outcome in newly diagnosed multiple myeloma. Hematol Oncol 2023; 41:912-921. [PMID: 37452600 DOI: 10.1002/hon.3208] [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: 03/19/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Until now, next generation sequencing (NGS) data has not been incorporated into any prognostic stratification of multiple myeloma (MM) and no therapeutic considerations are based upon it. In this work, we correlated NGS data with (1) therapy response and survival parameters in newly diagnosed multiple myeloma, treated by VRd * and (2) MM disease stage: newly diagnosed multiple myeloma (ndMM) versus relapsed and/or refractory (relapsed/refractory multiple myeloma). We analyzed 126 patients, with ndMM and relapsed refractory multiple myeloma (rrMM), treated at the University Hospital of Bern (Inselspital). Next generation sequencing was performed on bone marrow, as part of routine diagnostics. The NGS panel comprised eight genes CCND1, DIS3, EGR1, FAM46C (TENT5C), FGFR3, PRDM1, TP53, TRAF3 and seven hotspots in BRAF, IDH1, IDH2, IRF4, KRAS, NRAS. The primary endpoint was complete remission (CR) after VRd in ndMM, in correlation with mutational profile. Mutational load was generally higher in rrMM, with more frequently mutated TP53: 11/87 (13%) in ndMM versus 9/11 (81%) in rrMM (OR 0.0857, p = 0.0007). In ndMM, treated by VRd, mutations in MAPK-pathway members (NRAS, KRAS or BRAF) were associated with reduced probability of CR (21/38, 55%), as compared with wild type NRAS, KRAS or BRAF (34/40, 85%; OR 0.2225, p = 0.006). NRAS c.181C > A (p.Q61K) as a single mutation event showed a trend to reduced probability of achieving CR (OR 0.0912, p = 0.0247). Activation of MAPK pathway via mutated NRAS, KRAS and BRAF genes seems to have a negative impact on outcome in ndMM patients receiving VRd therapy. VRd* - bortezomib (Velcade®), lenalidomide (Revlimid®) and dexamethasone.
Collapse
Affiliation(s)
- Camille Perroud
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of Internal Medicine, Hôpital Cantonal Fribourgeois HFR, Fribourg, Switzerland
| | - Dario Thurian
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of Internal Medicine, Spital Thun STS AG, Thun, Switzerland
| | - Martin Andres
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Arnaud Künzi
- Clinical Trials Unit, University of Bern, Bern, Switzerland
| | - Gertrud Wiedemann
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Sacha Zeerleder
- Department of Hematology, Kantonsspital Luzern and University of Bern, Luzern, Switzerland
| | - Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Thomas Pabst
- Department of Clinical Oncology, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Naomi Porret
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Ekaterina Rebmann
- Department of Hematology and Central Hematology Laboratory, Inselspital, University Hospital of Bern, Bern, Switzerland
- Department of Oncology-Hematology, Hospital of Neuchâtel (RHNe), Neuchâtel, Switzerland
| |
Collapse
|
52
|
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
|
53
|
Wang P, Jin SY. Meta-analysis of the efficacy and safety of daratumumab in the treatment of multiple myeloma. World J Clin Cases 2023; 11:7091-7100. [PMID: 37946760 PMCID: PMC10631397 DOI: 10.12998/wjcc.v11.i29.7091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/22/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The treatment of multiple myeloma has significantly progressed over the past half-century. The purpose of this study was to perform a systematic review and meta-analysis in order to explore the efficacy and safety of daratumumab in treating multiple myeloma. AIM To explore the efficacy and safety of daratumumab in treating multiple myeloma. METHODS A systematic literature search was performed using Chinese and English databases, including the China National Knowledge Infrastructure, Wanfang, China Biology Medicine, VIP, the Cochrane Library, Embase, and PubMed. The search encompassed studies in treating multiple myeloma with daratumumab, spanning from the inception of the database to June 2023. Revman 5.1 software was used for analysis. RESULTS Our analysis included eight English articles and one Chinese article of high quality. The meta-analysis results indicated that compared to other therapies, daratumumab could improve the overall response rate (ORR) [odds ratio (OR) = 2.67, 95% confidence interval (CI) = 2.01, 3.53, Z = 6.85, P < 0.00001], complete remission (CR) (OR = 2.87, 95%CI = 2.16, 3.83, Z = 7.23, P < 0.00001) and progression-free survival (PFS) time (hazard ratio = 0.48, 95%CI = 0.38,0.60, Z = 6.54, P < 0.00001) in patients with multiple myeloma. These differences were statistically significant. Additionally, these results suggested that daratumumab increases the risk of neutropenia and thrombocytopenia with minimal effect on the incidences of anemia and upper respiratory tract infections. CONCLUSION Daratumumab can improve ORR, CR rate, and PFS in patients with multiple myeloma. It also increases the risk of neutropenia and thrombocytopenia, necessitating careful monitoring during its clinical application.
Collapse
Affiliation(s)
- Pei Wang
- Department of Hematology, Yanbian University Hospital, Yanji 133000, Jilin Province, China
| | - Sheng-Yu Jin
- Department of Hematology, Yanbian University Hospital, Yanji 133000, Jilin Province, China
| |
Collapse
|
54
|
Boyle EM, Blaney P, Stoeckle JH, Wang Y, Ghamlouch H, Gagler D, Braunstein M, Williams L, Tenenbaum A, Siegel A, Chen X, Varma G, Avigan J, Li A, Jinsi M, Kaminetzsky D, Arbini A, Montes L, Corre J, Rustad EH, Landgren O, Maura F, Walker BA, Bauer M, Bruno B, Tsirigos A, Davies FE, Morgan GJ. Multiomic Mapping of Acquired Chromosome 1 Copy-Number and Structural Variants to Identify Therapeutic Vulnerabilities in Multiple Myeloma. Clin Cancer Res 2023; 29:3901-3913. [PMID: 37449980 PMCID: PMC12166981 DOI: 10.1158/1078-0432.ccr-22-3209] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/27/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE Chromosome 1 (chr1) copy-number abnormalities (CNA) and structural variants (SV) are frequent in newly diagnosed multiple myeloma (NDMM) and are associated with a heterogeneous impact on outcomes, the drivers of which are largely unknown. EXPERIMENTAL DESIGN A multiomic approach comprising CRISPR, gene mapping of CNAs and SVs, methylation, expression, and mutational analysis was used to document the extent of chr1 molecular variants and their impact on pathway utilization. RESULTS We identified two distinct groups of gain(1q): focal gains associated with limited gene-expression changes and a neutral prognosis, and whole-arm gains, which are associated with substantial gene-expression changes, complex genetics, and an adverse prognosis. CRISPR identified a number of dependencies on chr1 but only limited variants associated with acquired CNAs. We identified seven regions of deletion, nine of gain, three of chromothripsis (CT), and two of templated insertion (TI), which contain a number of potential drivers. An additional mechanism involving hypomethylation of genes at 1q may contribute to the aberrant gene expression of a number of genes. Expression changes associated with whole-arm gains were substantial and gene set enrichment analysis identified metabolic processes, apoptotic resistance, signaling via the MAPK pathway, and upregulation of transcription factors as being key drivers of the adverse prognosis associated with these variants. CONCLUSIONS Multiple layers of genetic complexity impact the phenotype associated with CNAs on chr1 to generate its associated clinical phenotype. Whole-arm gains of 1q are the critically important prognostic group that deregulate multiple pathways, which may offer therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Eileen M Boyle
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Patrick Blaney
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, New York
| | - James H Stoeckle
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Yubao Wang
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Hussein Ghamlouch
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Dylan Gagler
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, New York
| | - Marc Braunstein
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Louis Williams
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
- Myeloma Group, Cleveland Clinic Foundation, Taussig Cancer Center, Cleveland, Ohio
| | - Avital Tenenbaum
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Ariel Siegel
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Xiaoyi Chen
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Gaurav Varma
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Jason Avigan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Alexander Li
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Monica Jinsi
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - David Kaminetzsky
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Arnaldo Arbini
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | | | - Jill Corre
- Unit for Genomics in Myeloma, Institut Universitaire du Cancer de Toulouse-Oncopole, University Hospital, Toulouse; Centre de Recherche en Cancérologie de Toulouse, Institut National de la Santé et de la Recherche Médicale U1037, Toulouse, France
| | - Even H Rustad
- Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ola Landgren
- Myeloma Service, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Francesco Maura
- Myeloma Service, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Brian A Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Department of Hematology Oncology, Indiana University, Indianapolis, Indiana
| | - Michael Bauer
- Department of Biomedical Informatics (DBMI), UAMS, Little-Rock, Arkansas
| | - Benedetto Bruno
- Department of Hematology, Azienda Ospedaliera Citta della Salute e della Scienza di Torino, Piemonte, Italy
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, New York
| | - Faith E Davies
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Gareth J Morgan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| |
Collapse
|
55
|
Giguère A, Raymond-Bouchard I, Collin V, Claveau JS, Hébert J, LeBlanc R. Optical Genome Mapping Reveals the Complex Genetic Landscape of Myeloma. Cancers (Basel) 2023; 15:4687. [PMID: 37835381 PMCID: PMC10571866 DOI: 10.3390/cancers15194687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/15/2023] [Accepted: 09/17/2023] [Indexed: 10/15/2023] Open
Abstract
Fluorescence in situ hybridization (FISH) on enriched CD138 plasma cells is the standard method for identification of clinically relevant genetic abnormalities in multiple myeloma. However, FISH is a targeted analysis that can be challenging due to the genetic complexity of myeloma. The aim of this study was to evaluate the potential of optical genome mapping (OGM) to detect clinically significant cytogenetic abnormalities in myeloma and to provide larger pangenomic information. OGM and FISH analyses were performed on CD138-purified cells of 20 myeloma patients. OGM successfully detected structural variants (SVs) (IGH and MYC rearrangements), copy number variants (CNVs) (17p/TP53 deletion, 1p deletion and 1q gain/amplification) and aneuploidy (gains of odd-numbered chromosomes, monosomy 13) classically expected with myeloma and led to a 30% increase in prognosis yield at our institution when compared to FISH. Despite challenges in the interpretation of OGM calls for CNV and aneuploidy losses in non-diploid genomes, OGM has the potential to replace FISH as the standard of care analysis in clinical settings and to efficiently change how we identify prognostic and predictive markers for therapies in the future. To our knowledge, this is the first study highlighting the feasibility and clinical utility of OGM in myeloma.
Collapse
Affiliation(s)
- Amélie Giguère
- Cytogenetics Laboratory, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada; (I.R.-B.); (V.C.); (J.H.)
| | - Isabelle Raymond-Bouchard
- Cytogenetics Laboratory, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada; (I.R.-B.); (V.C.); (J.H.)
| | - Vanessa Collin
- Cytogenetics Laboratory, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada; (I.R.-B.); (V.C.); (J.H.)
| | - Jean-Sébastien Claveau
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC H1T 2M4, Canada; (J.-S.C.); (R.L.)
| | - Josée Hébert
- Cytogenetics Laboratory, Maisonneuve-Rosemont Hospital, Montreal, QC H1T 2M4, Canada; (I.R.-B.); (V.C.); (J.H.)
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC H1T 2M4, Canada; (J.-S.C.); (R.L.)
| | - Richard LeBlanc
- Division of Hematology, Oncology and Transplantation, Department of Medicine, Maisonneuve-Rosemont Hospital, Université de Montréal, Montreal, QC H1T 2M4, Canada; (J.-S.C.); (R.L.)
| |
Collapse
|
56
|
Zhu G, Jin L, Shen W, Zhao M, Liu N. Intratumor microbiota: Occult participants in the microenvironment of multiple myeloma. Biochim Biophys Acta Rev Cancer 2023; 1878:188959. [PMID: 37488050 DOI: 10.1016/j.bbcan.2023.188959] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/20/2023] [Indexed: 07/26/2023]
Abstract
More recently, microbiota was detected in several tumorous tissues including multiple myeloma (MM), but the roles of which is still under-studied as paucity of research on tumor biology. Moreover, we also detected the presence of microbiota in the bone marrow of patients with MM by 2bRAD-M sequencing technology, which is an incurable hematological malignancy characterized by accumulation of abnormal plasma cells in the bone marrow. However, the roles of intratumor microbiota in tumor disease remains poorly understood. In this review, we critically reviewed recent literature about microbiota in the tumorigenesis and progression of MM. Importantly, we proposed that the emergence of microbiota in the microenvironment of multiple myeloma may be attributed to microbial dysbiosis and impaired intestinal barrier, due to the increased prevalence of MM in patients with obesity and diabetes, of which the characteristic phenotype is gut microbial dysbiosis and impaired intestinal barrier. When the intestinal barrier is damaged, dysbiotic microbiota and their metabolites, as well as dysregulated immune cells, may participate in the reshaping of the local immune microenvironment, and play pivotal roles in the tumorigenesis and development of multiple myeloma, probably by migrating to the bone marrow microenvironment from intestine. We also discuss the emerging microbiological manipulation strategies to improve long-term outcomes of MM, as well as the prospective of the state-of-the-art techniques to advance our knowledge about the biological implication in the microbiome in MM.
Collapse
Affiliation(s)
- Gengjun Zhu
- Central Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Lifang Jin
- Department of Oncology and Hematology, The Second Hospital of Jilin University, Changchun, China
| | - Weizhang Shen
- Department of Oncology and Hematology, The Second Hospital of Jilin University, Changchun, China
| | - Meng Zhao
- Department of Oncology and Hematology, The Second Hospital of Jilin University, Changchun, China
| | - Ning Liu
- Central Laboratory, The Second Hospital of Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China.
| |
Collapse
|
57
|
Xie C, Zhong L, Luo J, Luo J, Wu Y, Zheng S, Jiang L, Zhang J, Shi Y. Identification of mutation gene prognostic biomarker in multiple myeloma through gene panel exome sequencing and transcriptome analysis in Chinese population. Comput Biol Med 2023; 163:107224. [PMID: 37406588 DOI: 10.1016/j.compbiomed.2023.107224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/06/2023] [Accepted: 06/30/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND The 5-year survival rate of multiple myeloma (MM) in China is less than 40%, with considerable individual heterogeneity. Gene mutations are important predictive biomarkers that influence MM treatment decision. The aim of our study was to uncover the clinical significance of mutated genes in MM in the Chinese population. METHODS Targeted exon panel sequencing was performed of 400 genes to detect the gene mutation status in plasma cells from 50 patients with MM. DAVID was used to explore the functions and pathways of mutated genes. Detection of mutant gene expression, prognosis and immune cell infiltration with GSE6477. GEO2R was utilized to identify differentially expressed genes (DEGs). Kaplan-Meier and CIBERSORT were applied to compare survival distributions and evaluate the gene expression associated with immune cell infiltration, respectively. RESULTS Mutations of 337 genes were identified in MM. The mutation types included SNP, INS, and DEL, but the dominant mutation type was SNP. Function and pathway analysis of mutant genes were performed to elucidate DNA modifications. We identified a total number of 660 downregulated and 587 upregulated genes from the GSE6477 dataset. Thirty-three common genes were present in both the mutant genes and DEGs. The functions and pathways of the mutated genes were enriched in myeloid cell differentiation, regulation of hemopoiesis, etc. Moreover, we found that the low expression of BCL6, BIRC3, HLA-DQA1, and VCAN was correlated with poor prognosis in MM. CONCLUSIONS The mutations and low expression of BCL6, BIRC3, HLA-DQA1, and VCAN were correlated with poor prognosis and immune cell infiltration in MM. This study is the first to reveal the spectrum of mutations in the Chinese population by the use of an NGS panel.
Collapse
Affiliation(s)
- Chunbao Xie
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ling Zhong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jiangrong Luo
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ji Luo
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yingmiao Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shuai Zheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
| | - Jianbo Zhang
- Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
58
|
Kegyes D, Gulei D, Drula R, Cenariu D, Tigu B, Dima D, Tanase A, Badelita S, Buzoianu AD, Ciurea S, Ghiaur G, Terpos E, Ciechanover A, Einsele H, Tomuleasa C. Proteasome inhibition in combination with immunotherapies: State-of-the-Art in multiple myeloma. Blood Rev 2023; 61:101100. [PMID: 37291017 DOI: 10.1016/j.blre.2023.101100] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023]
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder accounting for around 1.8% of all neoplastic diseases. Nowadays, clinicians have a broad arsenal of drugs at their disposal for the treatment of MM, such as proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, bispecific antibodies, CAR T-cell therapies and antibody-drug conjugates. In this paper we briefly highlight essential clinical elements relating to proteasome inhibitors, such as bortezomib, carfilzomib and ixazomib. Studies suggest that the early use of immunotherapy may improve outcomes significantly. Therefore, in our review we specifically focus on the combination therapy of proteasome inhibitors with novel immunotherapies and/or transplant. A high number of patients develop PI resistance. Thus, we also review new generation PIs, such as marizomib, oprozomib (ONX0912) and delanzomib (CEP-18770) and their combinations with immunotherapies.
Collapse
Affiliation(s)
- David Kegyes
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Rares Drula
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Diana Cenariu
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bogdan Tigu
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Alina Tanase
- Department of Hematology and Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Sorina Badelita
- Department of Hematology and Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Anca-Dana Buzoianu
- Department of Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Stefan Ciurea
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California Irvine, CA, United States
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Aaron Ciechanover
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Hermann Einsele
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine / Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj Napoca, Romania; Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
| |
Collapse
|
59
|
Petrilla C, Galloway J, Kudalkar R, Ismael A, Cottini F. Understanding DNA Damage Response and DNA Repair in Multiple Myeloma. Cancers (Basel) 2023; 15:4155. [PMID: 37627183 PMCID: PMC10453069 DOI: 10.3390/cancers15164155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy characterized by several genetic abnormalities, including chromosomal translocations, genomic deletions and gains, and point mutations. DNA damage response (DDR) and DNA repair mechanisms are altered in MM to allow for tumor development, progression, and resistance to therapies. Damaged DNA rarely induces an apoptotic response, given the presence of ataxia-telangiectasia mutated (ATM) loss-of-function or mutations, as well as deletions, mutations, or downregulation of tumor protein p53 (TP53) and tumor protein p73 (TP73). Moreover, DNA repair mechanisms are either hyperactive or defective to allow for rapid correction of the damage or permissive survival. Medications used to treat patients with MM can induce DNA damage, by either direct effects (mono-adducts induced by melphalan), or as a result of reactive oxygen species (ROS) production by proteasome inhibitors such as bortezomib. In this review, we will describe the mechanisms of DDR and DNA repair in normal tissues, the contribution of these pathways to MM disease progression and other phenotypes, and the potential therapeutic opportunities for patients with MM.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Cottini
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
60
|
Brownlie RJ, Kennedy R, Wilson EB, Milanovic M, Taylor CF, Wang D, Davies JR, Owston H, Adams EJ, Stephenson S, Caeser R, Gewurz BE, Giannoudis PV, Scuoppo C, McGonagle D, Hodson DJ, Tooze RM, Doody GM, Cook G, Westhead DR, Klein U. Cytokine receptor IL27RA is an NF-κB-responsive gene involved in CD38 upregulation in multiple myeloma. Blood Adv 2023; 7:3874-3890. [PMID: 36867577 PMCID: PMC10405202 DOI: 10.1182/bloodadvances.2022009044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/27/2023] [Indexed: 03/04/2023] Open
Abstract
Multiple myeloma (MM) shows constitutive activation of canonical and noncanonical nuclear factor κB (NF-κB) signaling via genetic mutations or tumor microenvironment (TME) stimulations. A subset of MM cell lines showed dependency for cell growth and survival on the canonical NF-κB transcription factor RELA alone, suggesting a critical role for a RELA-mediated biological program in MM pathogenesis. Here, we determined the RELA-dependent transcriptional program in MM cell lines and found the expression of the cell surface molecules interleukin-27 receptor-α (IL-27Rα) and the adhesion molecule JAM2 to be responsive to RELA at the messenger RNA and protein levels. IL-27Rα and JAM2 were expressed on primary MM cells at higher levels than on healthy long-lived plasma cells (PCs) in the bone marrow. IL-27 activated STAT1, and to a lesser extent STAT3, in MM cell lines and in PCs generated from memory B cells in an IL-21-dependent in vitro PC differentiation assay. Concomitant activity of IL-21 and IL-27 enhanced differentiation into PCs and increased the cell-surface expression of the known STAT target gene CD38. In accordance, a subset of MM cell lines and primary MM cells cultured with IL-27 upregulated CD38 cell-surface expression, a finding with potential implications for enhancing the efficacy of CD38-directed monoclonal antibody therapies by increasing CD38 expression on tumor cells. The elevated expression of IL-27Rα and JAM2 on MM cells compared with that on healthy PCs may be exploited for the development of targeted therapeutic strategies that modulate the interaction of MM cells with the TME.
Collapse
Affiliation(s)
- Rebecca J. Brownlie
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Ruth Kennedy
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Erica B. Wilson
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Maja Milanovic
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY
| | - Claire F. Taylor
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Dapeng Wang
- Leeds Omics, University of Leeds, Leeds, United Kingdom
| | - John R. Davies
- Bioinformatics Group, School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Heather Owston
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Emma J. Adams
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Sophie Stephenson
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Rebecca Caeser
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Peter V. Giannoudis
- Leeds Orthopaedic & Trauma Sciences, Leeds General Infirmary, and Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Claudio Scuoppo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Daniel J. Hodson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Reuben M. Tooze
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Gina M. Doody
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| | - Gordon Cook
- CRUK Clinical Trials Unit, Leeds Institute of Clinical Trial Research, University of Leeds, Leeds, United Kingdom
| | - David R. Westhead
- Bioinformatics Group, School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Ulf Klein
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James’s Hospital, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
61
|
Sklavenitis-Pistofidis R, Lightbody ED, Reidy M, Tsuji J, Aranha MP, Heilpern-Mallory D, Huynh D, Chong SJF, Hackett L, Haradhvala NJ, Wu T, Su NK, Berrios B, Alberge JB, Dutta A, Davids MS, Papaioannou M, Getz G, Ghobrial IM, Manier S. Systematic characterization of therapeutic vulnerabilities in Multiple Myeloma with Amp1q reveals increased sensitivity to the combination of MCL1 and PI3K inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551480. [PMID: 37577538 PMCID: PMC10418223 DOI: 10.1101/2023.08.01.551480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The development of targeted therapy for patients with Multiple Myeloma (MM) is hampered by the low frequency of actionable genetic abnormalities. Gain or amplification of chr1q (Amp1q) is the most frequent arm-level copy number gain in patients with MM, and it is associated with higher risk of progression and death despite recent advances in therapeutics. Thus, developing targeted therapy for patients with MM and Amp1q stands to benefit a large portion of patients in need of more effective management. Here, we employed large-scale dependency screens and drug screens to systematically characterize the therapeutic vulnerabilities of MM with Amp1q and showed increased sensitivity to the combination of MCL1 and PI3K inhibitors. Using single-cell RNA sequencing, we compared subclones with and without Amp1q within the same patient tumors and showed that Amp1q is associated with higher levels of MCL1 and the PI3K pathway. Furthermore, by isolating isogenic clones with different copy number for part of the chr1q arm, we showed increased sensitivity to MCL1 and PI3K inhibitors with arm-level gain. Lastly, we demonstrated synergy between MCL1 and PI3K inhibitors and dissected their mechanism of action in MM with Amp1q.
Collapse
Affiliation(s)
- Romanos Sklavenitis-Pistofidis
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elizabeth D. Lightbody
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Mairead Reidy
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Junko Tsuji
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Michelle P. Aranha
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daniel Heilpern-Mallory
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Daisy Huynh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J. F. Chong
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Liam Hackett
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nicholas J. Haradhvala
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ting Wu
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Nang K. Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Brianna Berrios
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jean-Baptiste Alberge
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Ankit Dutta
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Matthew S. Davids
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Maria Papaioannou
- Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Hematology Unit, 1st Internal Medicine Department, AHEPA University Hospital, Thessaloniki, Greece
| | - Gad Getz
- Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Irene M. Ghobrial
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Salomon Manier
- INSERM UMRS1277, CNRS UMR9020, Lille University, 59000, France
- Department of Hematology, CHU Lille, Lille University, 59000, France
| |
Collapse
|
62
|
Abstract
Multiple myeloma (MM) is the second most common form of blood cancer characterized by clonal expansion of malignant plasma cells within the bone marrow. MM is a complex, progressive, and highly heterogeneous malignancy, which occurs via a multistep transformation process involving primary and secondary oncogenic events. Recent advances in molecular techniques have further expanded our understanding of the mutational landscape, clonal composition, and dynamic evolution patterns of MM. The first part of this review describes the key oncogenic events involved in the initiation and progression of MM, together with their prognostic impact. The latter part highlights the most prominent findings concerning genomic aberrations promoted by gene expression profiling (GEP) and next-generation sequencing (NGS) in MM. This review provides a concise understanding of the molecular pathogenesis of the MM genome and the importance of adopting emerging molecular technology in future clinical management of MM.
Collapse
Affiliation(s)
- Ivyna Pau Ni Bong
- Hematology Unit, Cancer Research Center, Institute for Medical Research, National Institute of Health, Ministry of Health, Malaysia
| | - Ezalia Esa
- Hematology Unit, Cancer Research Center, Institute for Medical Research, National Institute of Health, Ministry of Health, Malaysia
| |
Collapse
|
63
|
McAvera R, Quinn J, Murphy P, Glavey S. Genetic Abnormalities in Extramedullary Multiple Myeloma. Int J Mol Sci 2023; 24:11259. [PMID: 37511018 PMCID: PMC10379577 DOI: 10.3390/ijms241411259] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Extramedullary multiple myeloma (or extramedullary disease, EMD) is an aggressive form of multiple myeloma (MM) that occurs when malignant plasma cells become independent of the bone marrow microenvironment. This may occur alongside MM diagnosis or in later stages of relapse and confers an extremely poor prognosis. In the era of novel agents and anti-myeloma therapies, the incidence of EMD is increasing, making this a more prevalent and challenging cohort of patients. Therefore, understanding the underlying mechanisms of bone marrow escape and EMD driver events is increasingly urgent. The role of genomics in MM has been studied extensively; however, much less is known about the genetic background of EMD. Recently there has been an increased focus on driver events for the establishment of distant EMD sites. Generally, high-risk cytogenetic abnormalities and gene signatures are associated with EMD, alongside mutations in RAS signalling pathways. More recently, changes in epigenetic regulation have also been documented, specifically the hypermethylation of DNA promoter regions. Therefore, the focus of this review is to summarize and discuss what is currently known about the genetic background of EMD in MM.
Collapse
Affiliation(s)
- Roisin McAvera
- Department of Pathology, Royal College of Surgeons in Ireland, D09 YD60 Dublin, Ireland
| | - John Quinn
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
- School of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Philip Murphy
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
| | - Siobhan Glavey
- Department of Pathology, Royal College of Surgeons in Ireland, D09 YD60 Dublin, Ireland
- Department of Haematology, Beaumont Hospital, D09 V2N0 Dublin, Ireland
| |
Collapse
|
64
|
Zhou P, Li W, Zuo S, Ma R, Yuan X, Zhu Z. Pathogenesis, clinical characteristics and personalized managements of multiple myeloma with chromosome 1 abnormalities. Leuk Lymphoma 2023; 64:1373-1388. [PMID: 37300424 DOI: 10.1080/10428194.2023.2216325] [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: 01/30/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023]
Abstract
Multiple myeloma (MM) is a biologically heterogeneous malignancy defined by the proliferation of monoclonal plasma cells. Despite the tremendous advancement in MM treatment over the past decades, relapse remains a major problem which is inevitable for most patients. In particular, a partial of patients with early relapse and poor outcomes are classified as a high-risk group. Apart from the clinical stage, genetic aberrations are now recognized as important prognostic factors for identifying high-risk patients. Chromosome 1 abnormalities (C1As), particularly 1q21 gain or amplification, have been identified as common genetic aberrations in patients with MM and are often considered unfavorable prognostic markers for progression-free survival and overall survival. However, more effective therapeutic approaches are still needed to overcome the negative impact of C1As. Therefore, we summarize the prevalence, pathogenesis, clinical significance and present therapeutic condition of C1As in MM, and attempt to conclude the precise and personalized management for patients with C1As.
Collapse
Affiliation(s)
- Pan Zhou
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Weiya Li
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Suqiong Zuo
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Rongjun Ma
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Xiaoli Yuan
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| | - Zunmin Zhu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, P.R. China
| |
Collapse
|
65
|
Rana PS, Goparaju K, Driscoll JJ. Shutting off the fuel supply to target metabolic vulnerabilities in multiple myeloma. Front Oncol 2023; 13:1141851. [PMID: 37361580 PMCID: PMC10285382 DOI: 10.3389/fonc.2023.1141851] [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: 01/10/2023] [Accepted: 05/18/2023] [Indexed: 06/28/2023] Open
Abstract
Pathways that govern cellular bioenergetics are deregulated in tumor cells and represent a hallmark of cancer. Tumor cells have the capacity to reprogram pathways that control nutrient acquisition, anabolism and catabolism to enhance their growth and survival. Tumorigenesis requires the autonomous reprogramming of key metabolic pathways that obtain, generate and produce metabolites from a nutrient-deprived tumor microenvironment to meet the increased bioenergetic demands of cancer cells. Intra- and extracellular factors also have a profound effect on gene expression to drive metabolic pathway reprogramming in not only cancer cells but also surrounding cell types that contribute to anti-tumor immunity. Despite a vast amount of genetic and histologic heterogeneity within and between cancer types, a finite set of pathways are commonly deregulated to support anabolism, catabolism and redox balance. Multiple myeloma (MM) is the second most common hematologic malignancy in adults and remains incurable in the vast majority of patients. Genetic events and the hypoxic bone marrow milieu deregulate glycolysis, glutaminolysis and fatty acid synthesis in MM cells to promote their proliferation, survival, metastasis, drug resistance and evasion of immunosurveillance. Here, we discuss mechanisms that disrupt metabolic pathways in MM cells to support the development of therapeutic resistance and thwart the effects of anti-myeloma immunity. A better understanding of the events that reprogram metabolism in myeloma and immune cells may reveal unforeseen vulnerabilities and advance the rational design of drug cocktails that improve patient survival.
Collapse
Affiliation(s)
- Priyanka S. Rana
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Immune Oncology Program, Case Comprehensive Cancer Center, Cleveland, OH, United States
| | - Krishna Goparaju
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - James J. Driscoll
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Immune Oncology Program, Case Comprehensive Cancer Center, Cleveland, OH, United States
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
66
|
Chen M, Jiang J, Hou J. Single-cell technologies in multiple myeloma: new insights into disease pathogenesis and translational implications. Biomark Res 2023; 11:55. [PMID: 37259170 PMCID: PMC10234006 DOI: 10.1186/s40364-023-00502-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by clonal proliferation of plasma cells. Although therapeutic advances have been made to improve clinical outcomes and to prolong patients' survival in the past two decades, MM remains largely incurable. Single-cell sequencing (SCS) is a powerful method to dissect the cellular and molecular landscape at single-cell resolution, instead of providing averaged results. The application of single-cell technologies promises to address outstanding questions in myeloma biology and has revolutionized our understanding of the inter- and intra-tumor heterogeneity, tumor microenvironment, and mechanisms of therapeutic resistance in MM. In this review, we summarize the recently developed SCS methodologies and latest MM research progress achieved by single-cell profiling, including information regarding the cancer and immune cell landscapes, tumor heterogeneities, underlying mechanisms and biomarkers associated with therapeutic response and resistance. We also discuss future directions of applying transformative SCS approaches with contribution to clinical translation.
Collapse
Affiliation(s)
- Mengping Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jinxing Jiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
67
|
Liu Z, Yang C, Liu X, Xu X, Zhao X, Fu R. Therapeutic strategies to enhance immune response induced by multiple myeloma cells. Front Immunol 2023; 14:1169541. [PMID: 37275861 PMCID: PMC10232766 DOI: 10.3389/fimmu.2023.1169541] [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: 02/19/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Multiple myeloma (MM)as a haematological malignancy is still incurable. In addition to the presence of somatic genetic mutations in myeloma patients, the presence of immunosuppressive microenvironment greatly affects the outcome of treatment. Although the discovery of immunotherapy makes it possible to break the risk of high toxicity and side effects of traditional chemotherapeutic drugs, there are still obstacles of ineffective treatment or disease recurrence. In this review, we discuss therapeutic strategies to further enhance the specific anti-tumor immune response by activating the immunogenicity of MM cells themselves. New ideas for future myeloma therapeutic approaches are provided.
Collapse
|
68
|
Wittner J, Schuh W. Krüppel-like factor 2: a central regulator of B cell differentiation and plasma cell homing. Front Immunol 2023; 14:1172641. [PMID: 37251374 PMCID: PMC10213221 DOI: 10.3389/fimmu.2023.1172641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/06/2023] [Indexed: 05/31/2023] Open
Abstract
The development of B cells, their activation and terminal differentiation into antibody-producing plasma cells are characterized by alternating phases of proliferation and quiescence that are controlled by complex transcriptional networks. The spatial and anatomical organization of B cells and plasma cells inside lymphoid organs as well as their migration within lymphoid structures and between organs are prerequisites for the generation and the maintenance of humoral immune responses. Transcription factors of the Krüppel-like family are critical regulators of immune cell differentiation, activation, and migration. Here, we discuss the functional relevance of Krüppel-like factor 2 (KLF2) for B cell development, B cell activation, plasma cell formation and maintenance. We elaborate on KLF2-mediated regulation of B cell and plasmablast migration in the context of immune responses. Moreover, we describe the importance of KLF2 for the onset and the progression of B cell-related diseases and malignancies.
Collapse
|
69
|
Liu Z, Jia Y, Yang C, Liu H, Shen H, Wang H, Fu R. Study on the Effect of EZH2 Inhibitor Combined with TIGIT Monoclonal Antibody against Multiple Myeloma Cells. Int J Mol Sci 2023; 24:ijms24108603. [PMID: 37239949 DOI: 10.3390/ijms24108603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
EZH2, a member of the polycomb repressive complex 2, induces trimethylation of the downstream gene at the histone three lysine 27 (H3K27me3) position to inhibit tumor cell proliferation. Here, we showed that the apoptosis rate and apoptotic protein expression increased after EZH2 inhibition, whereas key molecules of the NF-κB signaling pathway and the downstream target genes were inhibited. Additionally, the expression of CD155, a TIGIT high-affinity ligand in multiple myeloma (MM) cells, was decreased by the mTOR signaling pathway. Furthermore, the combination of EZH2 inhibitor and TIGIT monoclonal antibody blockade enhanced the anti-tumor effect of natural killer cells. In summary, the EZH2 inhibitor not only plays an anti-tumor role as an epigenetic drug, but also enhances the anti-tumor effect of the TIGIT monoclonal antibody by affecting the TIGIT-CD155 axis between NK cells and MM cells, thus providing new ideas and theoretical basis for the treatment of MM patients.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| | - Yue Jia
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| | - Chun Yang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| | - Hao Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, China
| |
Collapse
|
70
|
Schavgoulidze A, Talbot A, Perrot A, Cazaubiel T, Leleu X, Manier S, Buisson L, Mahéo S, Do Souto Ferreira L, Pavageau L, Hulin C, Marolleau JP, Voillat L, Belhadj K, Divoux M, Slama B, Brechignac S, Macro M, Stoppa AM, Sanhes L, Orsini-Piocelle F, Fontan J, Chretien ML, Demarquette H, Mohty M, Avet-Loiseau H, Corre J. Biallelic deletion of 1p32 defines ultra-high-risk myeloma, but monoallelic del(1p32) remains a strong prognostic factor. Blood 2023; 141:1308-1315. [PMID: 36375118 PMCID: PMC10163308 DOI: 10.1182/blood.2022017863] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 11/01/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
Cytogenetic abnormalities (CAs) are known to be the preponderant prognostic factor in multiple myeloma. Our team has recently developed a prognostic score based on 6 CAs, with which del(1p32) appears to be the second worst abnormality after del(17p). This study aimed to confirm the adverse effect of 1p32 deletion in patients with newly diagnosed multiple myeloma (NDMM). Among 2551 patients with newly diagnosed multiple myeloma, 11% were harboring del(1p32). Their overall survival (OS) was significantly inferior compared with patients without del(1p32) (median OS: 49 months vs 124 months). Likewise, progression-free survival was significantly shorter. More importantly, biallelic del(1p32) conferred a dramatically poorer prognosis than a monoallelic del(1p32) (median OS: 25 months vs 60 months). As expected, the OS of patients with del(1p32) significantly decreased when this abnormality was associated with other high-risk CAs [del(17p), t(4;14), or gain(1q)]. In the multivariate analysis, del(1p32) appeared as a negative prognostic factor; after adjustment for age and treatment, the risk of progression was 1.3 times higher among patients harboring del(1p32), and the risk of death was 1.9 times higher. At the dawn of risk-adapted treatment strategies, we have confirmed the adverse effect of del(1p32) in multiple myeloma and the relevance of its assessment at diagnosis.
Collapse
Affiliation(s)
- Anaïs Schavgoulidze
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| | - Alexis Talbot
- Hematology Department, Saint-Louis University Hospital, University of Paris, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Aurore Perrot
- Hematology Department, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse, France
| | - Titouan Cazaubiel
- Hematology Department, Bordeaux University Hospital, Bordeaux, France
| | - Xavier Leleu
- Hematology Department, Poitiers University Hospital, Poitiers, France
| | - Salomon Manier
- Hematology Department, Lille University Hospital, Lille, France
| | - Laure Buisson
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| | - Sabrina Mahéo
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| | - Laura Do Souto Ferreira
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| | - Luka Pavageau
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| | - Cyrille Hulin
- Hematology Department, Bordeaux University Hospital, Bordeaux, France
| | | | - Laurent Voillat
- Hematology Department, Chalon-sur-Saône Hospital, Chalon-sur-Saône, France
| | - Karim Belhadj
- Hematology Department, Créteil University Hospital, Créteil, France
| | - Marion Divoux
- Hematology Department, Nancy University Hospital, Nancy, France
| | - Borhane Slama
- Hematology Department, Avignon Hospital, Avignon, France
| | | | - Margaret Macro
- Hematology Department, Caen University Hospital, Caen, France
| | | | - Laurence Sanhes
- Hematology Department, Perpignan Hospital, Perpignan, France
| | | | - Jean Fontan
- Hematology Department, Besançon University Hospital, Besançon, France
| | | | | | - Mohamad Mohty
- Hematology Department, Saint-Antoine University Hospital, Paris, France
| | - Hervé Avet-Loiseau
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| | - Jill Corre
- Unit for Genomics in Myeloma, University Hospital IUC-Oncopole and Toulouse Cancer Research Center-Oncopole, Toulouse University, INSERM UMR1037, Toulouse, France
| |
Collapse
|
71
|
Winkler W, Farré Díaz C, Blanc E, Napieczynska H, Langner P, Werner M, Walter B, Wollert-Wulf B, Yasuda T, Heuser A, Beule D, Mathas S, Anagnostopoulos I, Rosenwald A, Rajewsky K, Janz M. Mouse models of human multiple myeloma subgroups. Proc Natl Acad Sci U S A 2023; 120:e2219439120. [PMID: 36853944 PMCID: PMC10013859 DOI: 10.1073/pnas.2219439120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/26/2023] [Indexed: 03/01/2023] Open
Abstract
Multiple myeloma (MM), a tumor of germinal center (GC)-experienced plasma cells, comprises distinct genetic subgroups, such as the t(11;14)/CCND1 and the t(4;14)/MMSET subtype. We have generated genetically defined, subgroup-specific MM models by the GC B cell-specific coactivation of mouse Ccnd1 or MMSET with a constitutively active Ikk2 mutant, mimicking the secondary NF-κB activation frequently seen in human MM. Ccnd1/Ikk2ca and MMSET/Ikk2ca mice developed a pronounced, clonally restricted plasma cell outgrowth with age, accompanied by serum M spikes, bone marrow insufficiency, and bone lesions. The transgenic plasma cells could be propagated in vivo and showed distinct transcriptional profiles, resembling their human MM counterparts. Thus, we show that targeting the expression of genes involved in MM subgroup-specific chromosomal translocations into mouse GC B cells translates into distinct MM-like diseases that recapitulate key features of the human tumors, opening the way to a better understanding of the pathogenesis and therapeutic vulnerabilities of different MM subgroups.
Collapse
Affiliation(s)
- Wiebke Winkler
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| | - Carlota Farré Díaz
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| | - Eric Blanc
- Core Unit Bioinformatics, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin10117, Germany
| | - Hanna Napieczynska
- Animal Phenotyping, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
| | - Patrick Langner
- Animal Phenotyping, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
| | - Marvin Werner
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| | - Barbara Walter
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| | - Brigitte Wollert-Wulf
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| | - Tomoharu Yasuda
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
| | - Arnd Heuser
- Animal Phenotyping, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin10117, Germany
| | - Stephan Mathas
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| | - Ioannis Anagnostopoulos
- Institute of Pathology, Universität Würzburg and Comprehensive Cancer Centre Mainfranken, Würzburg97080, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Universität Würzburg and Comprehensive Cancer Centre Mainfranken, Würzburg97080, Germany
| | - Klaus Rajewsky
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
| | - Martin Janz
- Biology of Malignant Lymphomas, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin13125, Germany
- Experimental and Clinical Research Center, a cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Berlin13125, Germany
- Hematology, Oncology and Cancer Immunology, Charité – Universitätsmedizin Berlin, Berlin13125, Germany
| |
Collapse
|
72
|
Andreozzi F, Dragani M, Quivoron C, Le Bras F, Assi T, Danu A, Belhadj K, Lazarovici J, Cotteret S, Bernard OA, Ribrag V, Michot JM. Precision Medicine Approach Based on Molecular Alterations for Patients with Relapsed or Refractory Multiple Myeloma: Results from the MM-EP1 Study. Cancers (Basel) 2023; 15:cancers15051508. [PMID: 36900299 PMCID: PMC10001403 DOI: 10.3390/cancers15051508] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Despite that cytogenetic and molecular analysis of tumor cells can rapidly identify recurring molecular abnormalities, no personalized therapy is currently available in the setting of relapsed/refractory multiple myeloma (r/r MM). METHODS MM-EP1 is a retrospective study aimed at comparing a personalized molecular-oriented (MO) versus a non-molecular-oriented (no-MO) approach in r/r MM. Actionable molecular targets and their associated therapies were the BRAF V600E mutation and BRAF inhibitors; t(11;14)(q13;q32) and BCL2 inhibitors; and t(4;14)(p16;q32) with FGFR3 fusion/rearrangements and FGFR3 inhibitors. RESULTS One hundred three highly pretreated r/r MM patients with a median age of 67 years (range 44-85) were included. Seventeen (17%) patients were treated using an MO approach with BRAF inhibitors (vemurafenib or dabrafenib, n = 6), BCL2 inhibitor (venetoclax, n = 9), or FGFR3 inhibitor (erdafitinib, n = 2). Eighty-six (86%) patients received non-MO therapies. Overall response rate was 65% in MO patients versus 58% in the non-MO group (p = 0.053). Median PFS and OS were 9 and 6 months (HR = 0.96; CI95 = 0.51-1.78; p = 0.88) and 26 and 28 months (HR = 0.98; CI95 = 0.46-2.12; p = 0.98), respectively, in MO and no-MO patients. CONCLUSION Despite the low number of patients treated with an MO approach, this study highlights the strengths and weakness of a molecular-targeted approach for the treatment of multiple myeloma. Widespread biomolecular techniques and improvement of precision medicine treatment algorithms could improve selection for precision medicine in myeloma.
Collapse
Affiliation(s)
- Fabio Andreozzi
- Département d’Innovation Thérapeutique et d’Essais Précoces, Gustave Roussy, 94805 Villejuif, France
- Hematology Department, Institute Jules Bordet, 1070 Bruxelles, Belgium
- Correspondence:
| | - Matteo Dragani
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
| | - Cyril Quivoron
- Translational Research Hematological Laboratory, Gustave Roussy, 94805 Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| | - Fabien Le Bras
- Hematology Department, Assistance Publique Hôpitaux de Paris, Hôpital Henri Mondor, 94000 Créteil, France
| | - Tarek Assi
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
| | - Alina Danu
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
| | - Karim Belhadj
- Hematology Department, Assistance Publique Hôpitaux de Paris, Hôpital Henri Mondor, 94000 Créteil, France
| | | | - Sophie Cotteret
- Department of Medical Biology and Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Olivier A. Bernard
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| | - Vincent Ribrag
- Département d’Innovation Thérapeutique et d’Essais Précoces, Gustave Roussy, 94805 Villejuif, France
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
- Translational Research Hematological Laboratory, Gustave Roussy, 94805 Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| | - Jean-Marie Michot
- Département d’Innovation Thérapeutique et d’Essais Précoces, Gustave Roussy, 94805 Villejuif, France
- Hematology Department, Gustave Roussy, 94805 Villejuif, France
- INSERM U1170, Université Paris-Saclay, Gustave Roussy, 94805 Villejuif, France
| |
Collapse
|
73
|
Ohguchi Y, Ohguchi H. DIS3: The Enigmatic Gene in Multiple Myeloma. Int J Mol Sci 2023; 24:ijms24044079. [PMID: 36835493 PMCID: PMC9958658 DOI: 10.3390/ijms24044079] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/01/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Recent studies have revealed the genetic aberrations involved in the initiation and progression of various cancers, including multiple myeloma (MM), via next-generation sequencing analysis. Notably, DIS3 mutations have been identified in approximately 10% of patients with MM. Moreover, deletions of the long arm of chromosome 13, that includes DIS3, are present in approximately 40% of patients with MM. Regardless of the high incidence of DIS3 mutations and deletions, their contribution to the pathogenesis of MM has not yet been determined. Herein, we summarize the molecular and physiological functions of DIS3, focusing on hematopoiesis, and discuss the characteristics and potential roles of DIS3 mutations in MM. Recent findings highlight the essential roles of DIS3 in RNA homeostasis and normal hematopoiesis and suggest that the reduced activity of DIS3 may be involved in myelomagenesis by increasing genome instability.
Collapse
Affiliation(s)
- Yasuyo Ohguchi
- Division of Disease Epigenetics, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Hiroto Ohguchi
- Division of Disease Epigenetics, Institute of Resource Development and Analysis, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
74
|
Mithraprabhu S, Reynolds J, Turner R, Quach H, Horvath N, Kerridge I, Kalff A, Bergin K, Hocking J, Yuen F, Khong T, Durie BM, Spencer A. Circulating tumour DNA analysis predicts relapse and improves risk stratification in primary refractory multiple myeloma. Blood Cancer J 2023; 13:25. [PMID: 36781844 PMCID: PMC9925790 DOI: 10.1038/s41408-023-00796-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/15/2023] Open
Affiliation(s)
- Sridurga Mithraprabhu
- Australian Centre for Blood Diseases, Alfred Health-Monash University, Melbourne, VIC, Australia. .,Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia.
| | - John Reynolds
- Australian Centre for Blood Diseases, Alfred Health-Monash University, Melbourne, VIC, Australia.,Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Rose Turner
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Hang Quach
- St.Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia
| | | | - Ian Kerridge
- Royal North Shore Hospital, Sydney, NSW, Australia
| | - Anna Kalff
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Krystal Bergin
- Australian Centre for Blood Diseases, Alfred Health-Monash University, Melbourne, VIC, Australia.,Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Jay Hocking
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Flora Yuen
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Tiffany Khong
- Australian Centre for Blood Diseases, Alfred Health-Monash University, Melbourne, VIC, Australia.,Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia
| | - Brian M Durie
- Cedars-Sinai Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Andrew Spencer
- Australian Centre for Blood Diseases, Alfred Health-Monash University, Melbourne, VIC, Australia. .,Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
75
|
Jung J, Gokhale S, Xie P. TRAF3: A novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes. Front Oncol 2023; 13:1081253. [PMID: 36776285 PMCID: PMC9911533 DOI: 10.3389/fonc.2023.1081253] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Mitochondria, the organelle critical for cell survival and metabolism, are exploited by cancer cells and provide an important therapeutic target in cancers. Mitochondria dynamically undergo fission and fusion to maintain their diverse functions. Proteins controlling mitochondrial fission and fusion have been recognized as essential regulators of mitochondrial functions, mitochondrial quality control, and cell survival. In a recent proteomic study, we identified the key mitochondrial fission factor, MFF, as a new interacting protein of TRAF3, a known tumor suppressor of multiple myeloma and other B cell malignancies. This interaction recruits the majority of cytoplasmic TRAF3 to mitochondria, allowing TRAF3 to regulate mitochondrial morphology, mitochondrial functions, and mitochondria-dependent apoptosis in resting B lymphocytes. Interestingly, recent transcriptomic, metabolic and lipidomic studies have revealed that TRAF3 also vitally regulates multiple metabolic pathways in B cells, including phospholipid metabolism, glucose metabolism, and ribonucleotide metabolism. Thus, TRAF3 emerges as a novel regulator of mitochondrial physiology and metabolic pathways in B lymphocytes and B cell malignancies. Here we review current knowledge in this area and discuss relevant clinical implications.
Collapse
Affiliation(s)
- Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
76
|
Frontzek F, Hailfinger S, Lenz G. Plasmablastic lymphoma: from genetics to treatment. Leuk Lymphoma 2022; 64:799-807. [PMID: 36577021 DOI: 10.1080/10428194.2022.2162341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Plasmablastic lymphoma (PBL) represents a rare distinct lymphoma entity with plasmablastic morphology and plasmacytic immunophenotype that is characterized by an aggressive clinical course. Standard chemotherapeutic regimens often remain insufficient to cure affected patients. Recently, comprehensive molecular analyses of large cohorts of primary PBL samples have revealed the mutational landscape as well as the pattern of copy number alterations of this rare lymphoma subtype. Identification of recurrent aberrations affecting the JAK-STAT, RAS-RAF, NOTCH, IRF4, and MYC signaling pathways drive the molecular pathogenesis of PBL and hold great potential for novel targeted therapeutic approaches.
Collapse
Affiliation(s)
- Fabian Frontzek
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Stephan Hailfinger
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| |
Collapse
|
77
|
Evaluation of Genes and Molecular Pathways Involved in the Progression of Monoclonal Gammopathy of Undetermined Significance (MGUS) to Multiple Myeloma: A Systems Biology Approach. Mol Biotechnol 2022:10.1007/s12033-022-00634-6. [DOI: 10.1007/s12033-022-00634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
|
78
|
Testa S, Kumar J, Goodell AJ, Zehnder JL, Alexander KM, Sidana S, Arai S, Witteles RM, Liedtke M. Prevalence, mutational spectrum and clinical implications of clonal hematopoiesis of indeterminate potential in plasma cell dyscrasias. Semin Oncol 2022; 49:465-475. [PMID: 36503855 DOI: 10.1053/j.seminoncol.2022.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is common both in healthy individuals and patients with hematological cancers. Recent studies have showed worse prognosis for patients with multiple myeloma (MM) and non-Hodgkin lymphoma undergoing stem cell transplant, that have concomitant presence of CHIP. Data regarding the clinical and biological role of CHIP in plasma cell dyscrasias (PCDs) is rapidly increasing. However, the prevalence and prognostic implication of CHIP in patients with MM outside of the transplant setting, and in those with other more indolent PCDs remains elusive. Here we explored the prevalence and clinical implications of CHIP detected through next-generation sequencing in 209 patients with PCDs including MM, light chain (AL) amyloidosis (ALA), monoclonal gammopathy of undetermined significance (MGUS), and smoldering multiple myeloma (SMM). To avoid attributing the mutations to the plasma cell clone, CHIP was defined as the presence of DNMT3A, TET2, or ASXL1 mutations in the peripheral blood or bone marrow (DTA-CH). The prevalence of DTA-CH was 19% in patients with PCDs, with no difference between each PCD. TET2 (23%) and DNMT3A (22%), were the most frequently mutated genes. DTA-CH correlated with older age in MM (P = .001) and MGUS/SMM (P = 0.0007), as well as with coronary artery disease or congestive heart failure in MM (P = .03). DTA-CH did not predict worse OS or PFS in either MM or ALA, nor it predict higher risk of progression to MM in patients with MGUS/SMM. Our results overall further elucidate the prevalence and mutational spectrum of CHIP in PCDs, providing more information regarding the clinical relevance of CHIP in this patient population.
Collapse
Affiliation(s)
- Stefano Testa
- Department of Medicine, Stanford University, Stanford, CA, USA.
| | - Jyoti Kumar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex J Goodell
- Department of Medicine, Stanford University, Stanford, CA, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - James L Zehnder
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin M Alexander
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Surbhi Sidana
- Division of BMT and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Sally Arai
- Division of BMT and Cellular Therapy, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Ronald M Witteles
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michaela Liedtke
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
79
|
Zhang L, Zhang R, Wang J, Chen Y, Qiao C, Shi Q, Jin Y, Shen X, Li J, Chen L. Identification of clinical implications and potential prognostic models of chromatin regulator mutations in multiple myeloma. Clin Epigenetics 2022; 14:93. [PMID: 35870987 PMCID: PMC9308335 DOI: 10.1186/s13148-022-01314-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 07/16/2022] [Indexed: 12/29/2022] Open
Abstract
Abstract
Background
With the rapid development of next-generation sequencing (NGS) technologies, researchers are making efforts to reveal the genomic landscape of multiple myeloma (MM). However, the clinical significance of many mutations remains poorly defined due to the genetic heterogeneity of MM. To systematically explore the clinical implications of gene mutations and build practical prognostic models, we performed DNA sequencing in newly diagnosed MM patients.
Methods
MM cells were purified from bone marrow aspirates using CD138 microbeads and subjected to sequencing with a 387-gene Panel. Nomogram was developed using Cox’s proportional hazards model, and candidate variables were screened by stepwise regression. Internal validation was carried out by the bootstrap method.
Results
Between July 2016 and December 2020, a total of 147 patients were included in our study. We found patients with a higher mutational load had a significantly shorter progress-free survival (PFS) (19.0 vs. 32.0 months, P = 0.0098) and overall survival (OS) (3-year OS rates were 66.1% and 80.0%, P = 0.0290). Mutations in chromatin regulators (CRs) including KMT2C (14.3%), KMT2D (14.3%), EP300 (11.6%) and ARID gene family (31.3%) were highly frequent in newly diagnosed MM patients. Interestingly, proteins encoded by these genes could form a complex called KMT2C/D COMPASS (KCDCOMs). Patients with mutations of ARID gene family had a significantly shorter PFS (15.5 vs. 34.0 months, P = 0.0003) and OS (3-year OS rates were 64.9% and 81.0%, P = 0.0351) than patients without ARID gene mutations. Incorporating ARID gene mutations into the current staging system could successfully improve their prognostic performance. The PFS and OS nomogram models (including 1q21 copies, ARID gene mutations, extramedullary disease, mutational load and TP53 mutations) showed good predicting performance in both training and validation sets.
Conclusion
Our findings emphasized the importance of CRs mutations in newly diagnosed MM patients and indicated the mutations affecting KCDCOMs might promote the development of MM. High mutational load and harboring mutations in the ARID gene family were novel predictors of adverse prognosis in MM. Prognostic models based on gene mutations were commendably prognostic evaluation methods that could provide a reference for clinical practices.
Collapse
|
80
|
De Ramón C, Rojas EA, Cardona‐Benavides IJ, Mateos M, Corchete LA, Gutiérrez NC. Transcriptional signature of TP53 biallelic inactivation identifies a group of multiple myeloma patients without this genetic condition but with dismal outcome. Br J Haematol 2022; 199:344-354. [PMID: 35983648 PMCID: PMC9804640 DOI: 10.1111/bjh.18410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/25/2022] [Accepted: 08/05/2022] [Indexed: 01/09/2023]
Abstract
Biallelic inactivation of TP53 has been included in the definition of double-hit (DH) multiple myeloma (MM), which entails an ominous prognosis. However, this condition, or even the presence of high-risk cytogenetic abnormalities, cannot accurately capture the 15%-20% of the MM population with a median overall survival below 24 months. This prompted us to look for other MM patients who might have transcriptional characteristics similar to those with DH-TP53. In the present study, we analysed RNA-seq, whole-genome and whole-exome sequencing data from 660 newly diagnosed MM (NDMM) patients from the MMRF (Multiple Myeloma Research Foundation) CoMMpass study to characterize the transcriptional signature of TP53 double-hit (DH-TP53) MM. We found 78 genes that were exclusively deregulated in DH-TP53 patients. A score based on these genes identified a group of 50 patients who shared the same transcriptional profile (DH-TP53-like group) whose prognosis was particularly unfavourable [median overall survival (OS) < 2 years], despite not harbouring the biallelic inactivation of TP53. The prognostic value of the DH-TP53 score was externally validated using gene expression data from 850 NDMM patients analysed by microarrays. Furthermore, our DH-TP53 score refined the traditional prognostic stratification of MM patients according to the cytogenetic abnormalities and International Staging System (ISS).
Collapse
Affiliation(s)
- Cristina De Ramón
- Hematology DepartmentUniversity Hospital of Salamanca, IBSALSalamancaSpain,Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Elizabeta A. Rojas
- Hematology DepartmentUniversity Hospital of Salamanca, IBSALSalamancaSpain,Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Ignacio J. Cardona‐Benavides
- Hematology DepartmentUniversity Hospital of Salamanca, IBSALSalamancaSpain,Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Maria‐Victoria Mateos
- Hematology DepartmentUniversity Hospital of Salamanca, IBSALSalamancaSpain,Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233)SalamancaSpain
| | - Luis A. Corchete
- Hematology DepartmentUniversity Hospital of Salamanca, IBSALSalamancaSpain,Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233)SalamancaSpain
| | - Norma C. Gutiérrez
- Hematology DepartmentUniversity Hospital of Salamanca, IBSALSalamancaSpain,Cancer Research Center‐IBMCC (USAL‐CSIC)SalamancaSpain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC, CB16/12/00233)SalamancaSpain
| |
Collapse
|
81
|
Mejia Saldarriaga M, Darwiche W, Jayabalan D, Monge J, Rosenbaum C, Pearse RN, Niesvizky R, Bustoros M. Advances in the molecular characterization of multiple myeloma and mechanism of therapeutic resistance. Front Oncol 2022; 12:1020011. [PMID: 36387095 PMCID: PMC9646612 DOI: 10.3389/fonc.2022.1020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022] Open
Abstract
Recent insight in the genomic landscape of newly diagnosed multiple myeloma (NDMM) and its precursor conditions, monoclonal gammopathy of uncertain significance (MGUS), and smoldering myeloma have allowed the identification of patients with precursor conditions with a high risk of progression. These cases with "progressor" MGUS/SMM have a higher average mutation burden, have higher rates of mutations in specific genes such as MAPK, DNA repair, MYC, DIS3, and are enriched for specific mutational signatures when compared to non-progressors and are comparable to those found in NDMM. The highly preserved clonal heterogeneity seen upon progression of SMM, combined with the importance of these early variables, suggests that the identification of progressors based on these findings could complement and enhance the currently available clinical models based on tumor burden. Mechanisms leading to relapse/refractory multiple myeloma (RRMM) are of clinical interest given worse overall survival in this population. An Increased mutational burden is seen in patients with RRMM when compared to NDMM, however, there is evidence of branching evolution with many of these mutations being present at the subclonal level. Likewise, alterations in proteins associated with proteosome inhibitor and immunomodulatory drugs activity could partially explain clinical resistance to these agents. Evidence of chromosomal events leading to copy number changes is seen, with the presence of TP53 deletion, mutation, or a combination of both being present in many cases. Additional chromosomal events such as 1q gain and amplification may also interact and lead to resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mark Bustoros
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
82
|
Rosa-Rosa JM, Cuenca I, Medina A, Vázquez I, Sánchez-delaCruz A, Buenache N, Sánchez R, Jiménez C, Rosiñol L, Gutiérrez NC, Ruiz-Heredia Y, Barrio S, Oriol A, Martin-Ramos ML, Blanchard MJ, Ayala R, Ríos-Tamayo R, Sureda A, Hernández MT, de la Rubia J, Alkorta-Aranburu G, Agirre X, Bladé J, Mateos MV, Lahuerta JJ, San-Miguel JF, Calasanz MJ, Garcia-Sanz R, Martínez-Lopez J. NGS-Based Molecular Karyotyping of Multiple Myeloma: Results from the GEM12 Clinical Trial. Cancers (Basel) 2022; 14:cancers14205169. [PMID: 36291952 PMCID: PMC9601262 DOI: 10.3390/cancers14205169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/07/2022] [Accepted: 10/12/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Multiple Myeloma (MM) is considered an incurable chronic disease, which prognosis depends on the presence of different genomic alterations. To accomplish a complete molecular diagnosis in a single essay, we have designed and validated a capture-based NGS approach to reliably identify pathogenic mutations (SNVs and indels), genomic alterations (CNVs and chromosomic translocations), and IGH rearrangements. We have observed a good correlation of the results obtained using our capture panel with data obtained by both FISH and WES techniques. In this study, the molecular classification performed using our approach was significantly associated with the stratification and outcome of MM patients. Additionally, this panel has been proven to detect specific IGH rearrangements that could be used as biomarkers in patient follow-ups through minimal residual disease (MRD) assays. In conclusion, we think that MM patients could benefit from the use of this capture-based NGS approach with a more accurate, single-essay molecular diagnosis. Abstract Next-generation sequencing (NGS) has greatly improved our ability to detect the genomic aberrations occurring in multiple myeloma (MM); however, its transfer to routine clinical labs and its validation in clinical trials remains to be established. We designed a capture-based NGS targeted panel to identify, in a single assay, known genetic alterations for the prognostic stratification of MM. The NGS panel was designed for the simultaneous study of single nucleotide and copy number variations, insertions and deletions, chromosomal translocations and V(D)J rearrangements. The panel was validated using a cohort of 149 MM patients enrolled in the GEM2012MENOS65 clinical trial. The results showed great global accuracy, with positive and negative predictive values close to 90% when compared with available data from fluorescence in situ hybridization and whole-exome sequencing. While the treatments used in the clinical trial showed high efficacy, patients defined as high-risk by the panel had shorter progression-free survival (p = 0.0015). As expected, the mutational status of TP53 was significant in predicting patient outcomes (p = 0.021). The NGS panel also efficiently detected clonal IGH rearrangements in 81% of patients. In conclusion, molecular karyotyping using a targeted NGS panel can identify relevant prognostic chromosomal abnormalities and translocations for the clinical management of MM patients.
Collapse
Affiliation(s)
- Juan Manuel Rosa-Rosa
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
- Correspondence: (J.M.R.-R.); (J.M.-L.)
| | - Isabel Cuenca
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
| | - Alejandro Medina
- Unidad de Biología Molecular-HLA, Laboratorio de Hematología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Iria Vázquez
- Center for Applied Medical Research (CIMA) LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain
| | - Andrea Sánchez-delaCruz
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
| | - Natalia Buenache
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
| | - Ricardo Sánchez
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
| | - Cristina Jiménez
- Unidad de Biología Molecular-HLA, Laboratorio de Hematología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Laura Rosiñol
- Hospital Clinic de Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - Norma C. Gutiérrez
- Unidad de Biología Molecular-HLA, Laboratorio de Hematología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Yanira Ruiz-Heredia
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Santiago Barrio
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Albert Oriol
- Clinical Research Support Unit, Institut Català d’Oncologia, 08036 Barcelona, Spain
| | | | | | - Rosa Ayala
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
| | | | - Anna Sureda
- Institut Catalá d’Oncologia-l’Hospitalet, Institut de Investigació Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, 08908 Barcelona, Spain
| | | | | | - Gorka Alkorta-Aranburu
- Center for Applied Medical Research (CIMA) LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain
| | - Xabier Agirre
- Center for Applied Medical Research (CIMA) LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Universidad de Navarra, 31008 Pamplona, Spain
| | - Joan Bladé
- Hospital Clinic de Barcelona, 08036 Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
| | - María-Victoria Mateos
- Unidad de Biología Molecular-HLA, Laboratorio de Hematología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Juan-José Lahuerta
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Jesús F. San-Miguel
- Center for Applied Medical Research (CIMA) LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
- Instituto de Investigaciones Sanitarias de Navarra (IdiSNA), Universidad de Navarra, 31008 Pamplona, Spain
| | - María-José Calasanz
- Center for Applied Medical Research (CIMA) LAB Diagnostics, Universidad de Navarra, 31008 Pamplona, Spain
| | - Ramón Garcia-Sanz
- Unidad de Biología Molecular-HLA, Laboratorio de Hematología, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Joaquín Martínez-Lopez
- Hematology Department, Hospital 12 de Octubre, 28041 Madrid, Spain
- H12O–CNIO Hematological Malignancies Clinical Research Unit, Spanish National Cancer Research (CNIO), 29029 Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence: (J.M.R.-R.); (J.M.-L.)
| |
Collapse
|
83
|
Heestermans R, De Brouwer W, Maes K, Vande Broek I, Vaeyens F, Olsen C, Caljon B, De Becker A, Bakkus M, Schots R, Van Riet I. Liquid Biopsy-Derived DNA Sources as Tools for Comprehensive Mutation Profiling in Multiple Myeloma: A Comparative Study. Cancers (Basel) 2022; 14:cancers14194901. [PMID: 36230824 PMCID: PMC9563447 DOI: 10.3390/cancers14194901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is characterized by an expansion of plasma cells in the bone marrow (BM). The genetics of MM are highly complex with multiple mutations and genetic subpopulations of tumor cells that arise during the disease evolution, affecting prognosis and treatment response. Standard bone marrow DNA analysis requires an invasive sample collection and does not always reflect the complete mutation profile. Therefore, we examined the possibility to use peripheral blood-based liquid biopsies as an alternative DNA source for mutation profiling. By comparing DNA from circulating tumor cells with circulating tumor-derived vesicles and cell-free DNA (cfDNA), we found that the latter provided the best concordance with bone marrow DNA and also showed mutations derived from myeloma cell populations that were undetectable in bone marrow. Our comparative study indicates that cfDNA is the preferable circulating biomarker for genetic characterization in MM and can provide additional information compared to standard BM analysis. Abstract The analysis of bone marrow (BM) samples in multiple myeloma (MM) patients can lead to the underestimation of the genetic heterogeneity within the tumor. Blood-derived liquid biopsies may provide a more comprehensive approach to genetic characterization. However, no thorough comparison between the currently available circulating biomarkers as tools for mutation profiling in MM has been published yet and the use of extracellular vesicle-derived DNA for this purpose in MM has not yet been investigated. Therefore, we collected BM aspirates and blood samples in 30 patients with active MM to isolate five different DNA types, i.e., cfDNA, EV-DNA, BM-DNA and DNA isolated from peripheral blood mononucleated cells (PBMNCs-DNA) and circulating tumor cells (CTC-DNA). DNA was analyzed for genetic variants with targeted gene sequencing using a 165-gene panel. After data filtering, 87 somatic and 39 germline variants were detected among the 149 DNA samples used for sequencing. cfDNA showed the highest concordance with the mutation profile observed in BM-DNA and outperformed EV-DNA, CTC-DNA and PBMNCs-DNA. Of note, 16% of all the somatic variants were only detectable in circulating biomarkers. Based on our analysis, cfDNA is the preferable circulating biomarker for genetic characterization in MM and its combined use with BM-DNA allows for comprehensive mutation profiling in MM.
Collapse
Affiliation(s)
- Robbe Heestermans
- Department of Clinical Biology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Research Group Hematology and Immunology, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Wouter De Brouwer
- Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Research Group Hematology and Immunology, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ken Maes
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Isabelle Vande Broek
- Department of Oncology and Hematology, VITAZ, Moerlandstraat 1, 9100 Sint-Niklaas, Belgium
| | - Freya Vaeyens
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Catharina Olsen
- Clinical Sciences, Research Group Reproduction and Genetics, Centre for Medical Genetics, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Brussels Interuniversity Genomics High Throughput Core (BRIGHTcore), Vrije Universiteit Brussel (VUB), Université Libre de Bruxelles (ULB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Ben Caljon
- Brussels Interuniversity Genomics High Throughput Core (BRIGHTcore), Vrije Universiteit Brussel (VUB), Université Libre de Bruxelles (ULB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Ann De Becker
- Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Research Group Hematology and Immunology, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Marleen Bakkus
- Department of Clinical Biology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Research Group Hematology and Immunology, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Rik Schots
- Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Research Group Hematology and Immunology, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium
- Research Group Hematology and Immunology, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium
- Correspondence:
| |
Collapse
|
84
|
I-OPen: inferior outcomes of penta-refractory compared to penta-exposed multiple myeloma patients. Blood Cancer J 2022; 12:138. [PMID: 36151090 PMCID: PMC9508328 DOI: 10.1038/s41408-022-00733-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
|
85
|
Rebmann Chigrinova E, Porret NA, Andres M, Wiedemann G, Banz Y, Legros M, Pollak M, Oppliger Leibundgut E, Pabst T, Bacher U. Correlation of plasma cell assessment by phenotypic methods and molecular profiles by NGS in patients with plasma cell dyscrasias. BMC Med Genomics 2022; 15:203. [PMID: 36138464 PMCID: PMC9503268 DOI: 10.1186/s12920-022-01346-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 09/01/2022] [Indexed: 11/25/2022] Open
Abstract
Background Next-generation sequencing (NGS) detects somatic mutations in a high proportion of plasma cell dyscrasias (PCD), but is currently not integrated into diagnostic routine. We correlated NGS data with degree of bone marrow (BM) involvement by cytomorphology (BMC), histopathology (BMH), and multiparameter flow cytometry (MFC) in 90 PCD patients.
Methods Of the 90 patients the diagnoses comprised multiple myeloma (n = 77), MGUS (n = 7), AL-amyloidosis (n = 4) or solitary plasmocytoma (n = 2). The NGS panel included eight genes CCND1, DIS3, EGR1, FAM46C (TENT5C), FGFR3, PRDM1, TP53, TRAF3, and seven hotspots in BRAF, IDH1, IDH2, IRF4, KRAS, NRAS. Results Mutations were detected in 64/90 (71%) of cases. KRAS (29%), NRAS (16%) and DIS3 (16%) were most frequently mutated. At least one mutation/sample corresponded to a higher degree of BM involvement with a mean of 11% pathologic PC by MFC (range, 0.002–62%), and ~ 50% (3–100%) as defined by both BMC and BMH. Conclusions The probability of detecting a mutation by NGS in the BM was highest in samples with > 10% clonal PC by MFC, or > 20% PC by BMC/ BMH. We propose further evaluation of these thresholds as a practical cut-off for processing of samples by NGS at initial PCD diagnosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01346-1.
Collapse
Affiliation(s)
| | - Naomi A Porret
- Department of Hematology; Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin Andres
- Department of Hematology; Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gertrud Wiedemann
- Department of Hematology; Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Myriam Legros
- Center for Laboratory Medicine (ZLM), Inselspital, University of Bern, Bern, Switzerland
| | - Matthias Pollak
- Department of Hematology; Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Thomas Pabst
- Department of Medical Oncology, Inselspital, University of Bern, Bern, Switzerland
| | - Ulrike Bacher
- Department of Hematology; Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
86
|
Oncogenic RAS commandeers amino acid sensing machinery to aberrantly activate mTORC1 in multiple myeloma. Nat Commun 2022; 13:5469. [PMID: 36115844 PMCID: PMC9482638 DOI: 10.1038/s41467-022-33142-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
Oncogenic RAS mutations are common in multiple myeloma (MM), an incurable malignancy of plasma cells. However, the mechanisms of pathogenic RAS signaling in this disease remain enigmatic and difficult to inhibit therapeutically. We employ an unbiased proteogenomic approach to dissect RAS signaling in MM. We discover that mutant isoforms of RAS organize a signaling complex with the amino acid transporter, SLC3A2, and MTOR on endolysosomes, which directly activates mTORC1 by co-opting amino acid sensing pathways. MM tumors with high expression of mTORC1-dependent genes are more aggressive and enriched in RAS mutations, and we detect interactions between RAS and MTOR in MM patient tumors harboring mutant RAS isoforms. Inhibition of RAS-dependent mTORC1 activity synergizes with MEK and ERK inhibitors to quench pathogenic RAS signaling in MM cells. This study redefines the RAS pathway in MM and provides a mechanistic and rational basis to target this mode of RAS signaling. RAS mutations are commonly found in multiple myeloma (MM). Here, the authors show that oncogenic RAS mutations activate mTORC1 signalling in MM and combining mTORC1 and MEK/ERK inhibitors synergize to improve survival in preclinical models.
Collapse
|
87
|
Kaur G, Jena L, Gupta R, Farswan A, Gupta A, Sriram K. Correlation of changes in subclonal architecture with progression in the MMRF CoMMpass study. Transl Oncol 2022; 23:101472. [PMID: 35777247 PMCID: PMC9253848 DOI: 10.1016/j.tranon.2022.101472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 06/03/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous plasma cell proliferative disorder that arises from its premalignant precursor stages through a complex cascade of interactions between clonal mutations and co-evolving microenvironment. The temporo-spatial evolutionary trajectories of MM are established early during myelomatogenesis in precursor stages and retained in MM. Such molecular events impact subsequent disease progression and clinical outcomes. Identification of clonal sweeps of actionable gene targets in MM could reveal potential vulnerabilities that may exist in early stages and thus potentiate prognostication and customization of early therapeutic interventions. We have evaluated clonal evolution at multiple time points in 76 MM patients enrolled in the MMRF CoMMpass study. The major findings of this study are (a) MM progresses predominantly through branching evolution, (b) there is a heterogeneous spectrum of mutational landscapes that include unique actionable gene targets at diagnosis compared to progression, (c) unique clonal gains/ losses of mutant driver genes can be identified in patients with different cytogenetic aberrations, (d) there is a significant correlation between co-occurring oncogenic mutations/ co-occurring subclones e.g., with mutated TP53+SYNE1, NRAS+MAGI3, and anticorrelative dependencies between FAT3+FCGBP gene pairs. Such co-trajectories may synchronize molecular events of drug response, myelomatogenesis and warrant future studies to explore their potential for early prognostication and development of risk stratified personalized therapies in MM.
Collapse
Affiliation(s)
- Gurvinder Kaur
- Laboratory Oncology Unit, Dr. B. R.A. IRCH, AIIMS, New Delhi
| | - Lingaraja Jena
- Laboratory Oncology Unit, Dr. B. R.A. IRCH, AIIMS, New Delhi
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr. B. R.A. IRCH, AIIMS, New Delhi.
| | - Akanksha Farswan
- SBILab, Department of Electronics and Communication Engineering, IIIT, Delhi
| | - Anubha Gupta
- SBILab, Department of Electronics and Communication Engineering, IIIT, Delhi.
| | - K Sriram
- Department of Computational Biology & Centre for Computational Biology, IIIT, Delhi
| |
Collapse
|
88
|
Hergott CB, Kim AS. Molecular Diagnostic Testing for Hematopoietic Neoplasms: Linking Pathogenic Drivers to Personalized Diagnosis. Clin Lab Med 2022; 42:325-347. [PMID: 36150815 DOI: 10.1016/j.cll.2022.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Molecular diagnostics inhabit an increasingly central role in characterizing hematopoietic malignancies. This brief review summarizes the genomic targets important for many major categories of hematopoietic neoplasia by focusing on disease pathogenesis. In myeloid disease, recurrent mutations in key functional classes drive clonal hematopoiesis, on which additional variants can specify clinical presentation and accelerate progression. Lymphoblastic leukemias are frequently initiated by oncogenic fusions that block lymphoid maturation while, in concert with additional mutations, driving proliferation. The links between genetic aberrations and lymphoma patient outcomes have been clarified substantially through the clustering of genomic profiles. Finally, the addition of next-generation sequencing strategies to cytogenetics is refining risk stratification for plasma cell myeloma. In all categories, molecular diagnostics shed light on the unique mechanistic underpinnings of each individual malignancy, thereby empowering more rational, personalized care for these patients.
Collapse
Affiliation(s)
- Christopher B Hergott
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
89
|
Raimondi V, Iannozzi NT, Burroughs-Garcìa J, Toscani D, Storti P, Giuliani N. A personalized molecular approach in multiple myeloma: the possible use of RAF/RAS/MEK/ERK and BCL-2 inhibitors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:463-479. [PMID: 36071980 PMCID: PMC9446161 DOI: 10.37349/etat.2022.00095] [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: 03/15/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is a blood cancer that derives from plasma cells (PCs), which will accumulate in the bone marrow (BM). Over time, several drugs have been developed to treat this disease that is still uncurable. The therapies used to treat the disease target immune activity, inhibit proteasome activity, and involve the use of monoclonal antibodies. However, MM is a highly heterogeneous disease, in fact, there are several mutations in signaling pathways that are particularly important for MM cell biology and that are possible therapeutic targets. Indeed, some studies suggest that MM is driven by mutations within the rat sarcoma virus (RAS) signaling cascade, which regulates cell survival and proliferation. The RAS/proto-oncogene, serine/threonine kinase (RAF)/mitogen-activated extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK signaling pathway is deregulated in several cancers, for which drugs have been developed to inhibit these pathways. In addition to the signaling pathways, the disease implements mechanisms to ensure the survival and consequently a high replicative capacity. This strategy consists in the deregulation of apoptosis. In particular, some cases of MM show overexpression of anti-apoptotic proteins belonging to the B cell lymphoma 2 (BCL-2) family that represent a possible druggable target. Venetoclax is an anti-BCL-2 molecule used in hematological malignancies that may be used in selected MM patients based on their molecular profile. We focused on the possible effects in MM of off-label drugs that are currently used for other cancers with the same molecular characteristics. Their use, combined with the current treatments, could be a good strategy against MM.
Collapse
Affiliation(s)
- Vincenzo Raimondi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | | | | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;Hematology, “Azienda Ospedaliero-Universitaria di Parma”, 43126 Parma, Italy
| |
Collapse
|
90
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
91
|
Wang J, Hu Y, Hamidi H, Dos Santos C, Zhang J, Punnoose E, Li W. Immune microenvironment characteristics in multiple myeloma progression from transcriptome profiling. Front Oncol 2022; 12:948548. [PMID: 36033464 PMCID: PMC9413314 DOI: 10.3389/fonc.2022.948548] [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: 05/20/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Multiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow (BM). Despite the significant advances in treatment, relapsed and refractory MM has not yet been completely cured due to the immune dysfunction in the tumor microenvironment (TME). In this study, we analyzed the transcriptome data from patients with newly diagnosed (ND) and relapsed/refractory (R/R) MM to characterize differences in the TME and further decipher the mechanism of tumor progression in MM. We observed highly expressed cancer testis antigens and immune suppressive cell infiltration, such as Th2 and M2 cells, are associated with MM progression. Furthermore, the TGF-β signature contributes to the worse outcome of patients with R/R MM. Moreover, patients with ND MM could be classified into immune-low and immune-high phenotypes. Immune-high patients with higher IFN-g signatures are associated with MHC-II–mediated CD4+ T-cell response through CIITA stimulation. The baseline TME status could potentially inform new therapeutic choices for the ND MM who are ineligible for autologous stem cell transplantation and may help predict the response to CAR-T for patients with R/R MM. Our study demonstrates how integrating tumor transcriptome and clinical information to characterize MM immune microenvironment and elucidate potential mechanisms of tumor progression and immune evasion, which will provide insights into MM treatment selection.
Collapse
Affiliation(s)
- Jin Wang
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
| | - Yi Hu
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
| | - Habib Hamidi
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA, United States
| | - Cedric Dos Santos
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA, United States
| | - Jingyu Zhang
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
| | - Elizabeth Punnoose
- Oncology Biomarker Development, Genentech, Ltd., South San Francisco, CA, United States
| | - Wenjin Li
- Oncology Biomarker Development, Roche (China) Holding Ltd., Shanghai, China
- *Correspondence: Wenjin Li,
| |
Collapse
|
92
|
Gai D, Chen JR, Stewart JP, Nookaew I, Habelhah H, Ashby C, Sun F, Cheng Y, Li C, Xu H, Peng B, Garg TK, Schinke C, Thanendrarajan S, Zangari M, Chen F, Barlogie B, van Rhee F, Tricot G, Shaughnessy JD, Zhan F. CST6 suppresses osteolytic bone disease in multiple myeloma by blocking osteoclast differentiation. J Clin Invest 2022; 132:159527. [PMID: 35881476 PMCID: PMC9479617 DOI: 10.1172/jci159527] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
Osteolytic bone disease is a hallmark of multiple myeloma (MM). A significant fraction (~20%) of MM patients do not develop osteolytic lesions (OL). The molecular basis for the absence of bone disease in MM is not understood. We combined PET-CT and gene expression profiling (GEP) of purified bone marrow (BM) CD138+ MM cells from 512 newly diagnosed MM patients to reveal that elevated expression of cystatin M/E (CST6) was significantly associated with the absence of OL in MM. An enzyme-linked immunosorbent assay revealed a strong correlation between CST6 levels in BM serum/plasma and CST6 mRNA expression. Both recombinant CST6 protein and BM serum from patients with high CST6 significantly inhibited the activity of the osteoclast-specific protease cathepsin K, and blocked osteoclast differentiation and function. Recombinant CST6 inhibited bone destruction in ex vivo and in vivo myeloma models. Single cell RNA-sequencing identified that CST6 attenuates polarization of monocytes to osteoclast precursors. Furthermore, CST6 protein blocks osteoclast differentiation by suppressing cathepsin-mediated cleavage of NF-κB/p100 and TRAF3 following RANKL stimulation. Secretion by MM cells of CST6, an inhibitor of osteoclast differentiation and function, suppresses osteolytic bone disease in MM and probably other diseases associated with osteoclast-mediated bone loss.
Collapse
Affiliation(s)
- Dongzheng Gai
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Jin-Ran Chen
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - James P Stewart
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Intawat Nookaew
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Hasem Habelhah
- Department of Pathology, University of Iowa, Iowa City, United States of America
| | - Cody Ashby
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Fumou Sun
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Yan Cheng
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Can Li
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Hongwei Xu
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Bailu Peng
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Tarun K Garg
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Carolina Schinke
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Sharmilan Thanendrarajan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Maurizio Zangari
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Fangping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Bart Barlogie
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Frits van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Guido Tricot
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - John D Shaughnessy
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| | - Fenghuang Zhan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, United States of America
| |
Collapse
|
93
|
Ancos-Pintado R, Bragado-García I, Morales ML, García-Vicente R, Arroyo-Barea A, Rodríguez-García A, Martínez-López J, Linares M, Hernández-Sánchez M. High-Throughput CRISPR Screening in Hematological Neoplasms. Cancers (Basel) 2022; 14:3612. [PMID: 35892871 PMCID: PMC9329962 DOI: 10.3390/cancers14153612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023] Open
Abstract
CRISPR is becoming an indispensable tool in biological research, revolutionizing diverse fields of medical research and biotechnology. In the last few years, several CRISPR-based genome-targeting tools have been translated for the study of hematological neoplasms. However, there is a lack of reviews focused on the wide uses of this technology in hematology. Therefore, in this review, we summarize the main CRISPR-based approaches of high throughput screenings applied to this field. Here we explain several libraries and algorithms for analysis of CRISPR screens used in hematology, accompanied by the most relevant databases. Moreover, we focus on (1) the identification of novel modulator genes of drug resistance and efficacy, which could anticipate relapses in patients and (2) new therapeutic targets and synthetic lethal interactions. We also discuss the approaches to uncover novel biomarkers of malignant transformations and immune evasion mechanisms. We explain the current literature in the most common lymphoid and myeloid neoplasms using this tool. Then, we conclude with future directions, highlighting the importance of further gene candidate validation and the integration and harmonization of the data from CRISPR screening approaches.
Collapse
Affiliation(s)
- Raquel Ancos-Pintado
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - Irene Bragado-García
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - María Luz Morales
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
| | - Roberto García-Vicente
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
| | - Andrés Arroyo-Barea
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - Alba Rodríguez-García
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
| | - Joaquín Martínez-López
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
- Department of Medicine, Medicine School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain
| | - María Linares
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, CIBERONC, ES 28041 Madrid, Spain; (R.A.-P.); (M.L.M.); (R.G.-V.); (A.R.-G.); (J.M.-L.); (M.L.)
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| | - María Hernández-Sánchez
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, ES 28040 Madrid, Spain; (I.B.-G.); (A.A.-B.)
| |
Collapse
|
94
|
Salomon-Perzyński A, Barankiewicz J, Machnicki M, Misiewicz-Krzemińska I, Pawlak M, Radomska S, Krzywdzińska A, Bluszcz A, Stawiński P, Rydzanicz M, Jakacka N, Solarska I, Borg K, Spyra-Górny Z, Szpila T, Puła B, Grosicki S, Stokłosa T, Płoski R, Lech-Marańda E, Jakubikova J, Jamroziak K. Tracking Clonal Evolution of Multiple Myeloma Using Targeted Next-Generation DNA Sequencing. Biomedicines 2022; 10:biomedicines10071674. [PMID: 35884979 PMCID: PMC9313382 DOI: 10.3390/biomedicines10071674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 12/19/2022] Open
Abstract
Clonal evolution drives treatment failure in multiple myeloma (MM). Here, we used a custom 372-gene panel to track genetic changes occurring during MM progression at different stages of the disease. A tumor-only targeted next-generation DNA sequencing was performed on 69 samples sequentially collected from 30 MM patients. The MAPK/ERK pathway was mostly affected with KRAS mutated in 47% of patients. Acquisition and loss of mutations were observed in 63% and 37% of patients, respectively. Four different patterns of mutation evolution were found: branching-, mutation acquisition-, mutation loss- and a stable mutational pathway. Better response to anti-myeloma therapy was more frequently observed in patients who followed the mutation loss-compared to the mutation acquisition pathway. More than two-thirds of patients had druggable genes mutated (including cases of heavily pre-treated disease). Only 7% of patients had a stable copy number variants profile. Consequently, a redistribution in stages according to R-ISS between the first and paired samples (R-ISS″) was seen. The higher the R-ISS″, the higher the risk of MM progression and death. We provided new insights into the genetics of MM evolution, especially in heavily pre-treated patients. Additionally, we confirmed that redefining R-ISS at MM relapse is of high clinical value.
Collapse
Affiliation(s)
- Aleksander Salomon-Perzyński
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.S.-P.); (J.B.); (N.J.); (T.S.); (B.P.); (E.L.-M.)
| | - Joanna Barankiewicz
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.S.-P.); (J.B.); (N.J.); (T.S.); (B.P.); (E.L.-M.)
| | - Marcin Machnicki
- Department of Tumor Biology and Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (M.M.); (T.S.)
| | - Irena Misiewicz-Krzemińska
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (I.M.-K.); (M.P.)
| | - Michał Pawlak
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (I.M.-K.); (M.P.)
| | - Sylwia Radomska
- Molecular Biology Laboratory, Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (S.R.); (I.S.)
| | - Agnieszka Krzywdzińska
- Immunophenotyping Laboratory, Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland;
| | - Aleksandra Bluszcz
- Cytogenetic Laboratory, Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.B.); (K.B.)
| | - Piotr Stawiński
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (P.S.); (M.R.); (R.P.)
| | - Małgorzata Rydzanicz
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (P.S.); (M.R.); (R.P.)
| | - Natalia Jakacka
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.S.-P.); (J.B.); (N.J.); (T.S.); (B.P.); (E.L.-M.)
| | - Iwona Solarska
- Molecular Biology Laboratory, Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (S.R.); (I.S.)
| | - Katarzyna Borg
- Cytogenetic Laboratory, Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.B.); (K.B.)
| | - Zofia Spyra-Górny
- Department of Hematology and Cancer Prevention, Faculty od Health Sciences, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (Z.S.-G.); (S.G.)
| | - Tomasz Szpila
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.S.-P.); (J.B.); (N.J.); (T.S.); (B.P.); (E.L.-M.)
| | - Bartosz Puła
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.S.-P.); (J.B.); (N.J.); (T.S.); (B.P.); (E.L.-M.)
| | - Sebastian Grosicki
- Department of Hematology and Cancer Prevention, Faculty od Health Sciences, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (Z.S.-G.); (S.G.)
| | - Tomasz Stokłosa
- Department of Tumor Biology and Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (M.M.); (T.S.)
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (P.S.); (M.R.); (R.P.)
| | - Ewa Lech-Marańda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, 02-776 Warsaw, Poland; (A.S.-P.); (J.B.); (N.J.); (T.S.); (B.P.); (E.L.-M.)
| | - Jana Jakubikova
- Department of Tumor Immunology, Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, 84505 Bratislava, Slovakia;
| | - Krzysztof Jamroziak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-106 Warsaw, Poland
- Correspondence:
| |
Collapse
|
95
|
Dragoș ML, Ivanov IC, Mențel M, Văcărean-Trandafir IC, Sireteanu A, Titianu AA, Dăscălescu AS, Stache AB, Jitaru D, Gorgan DL. Prognostic Value of Association of Copy Number Alterations and Cell-Surface Expression Markers in Newly Diagnosed Multiple Myeloma Patients. Int J Mol Sci 2022; 23:7530. [PMID: 35886877 PMCID: PMC9318311 DOI: 10.3390/ijms23147530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma results from the clonal proliferation of abnormal plasma cells (PCs) in the bone marrow (BM). In this study, the cell surface expression markers (CD) on atypical PCs (detected by multiparametric flow cytometry (MFC)) were correlated with copy number alterations (CNAs) in the genome (detected by multiplex ligation-dependent probe amplification (MLPA)) to assess their impact on prognosis in newly diagnosed MM patients. Statistically significant results were obtained when different stages of PC maturation (classified based on CD19 and CD81 expression) were associated with CD117 expression and identified CNAs. In the intermediately differentiated PC group (CD19(-) CD81(+)), patients who didn't express CD117 had a lower median progression free survival (PFS) (p = 0.024). Moreover, within this group, patients with less than three adverse CNAs, which harbor CD117, had a better outcome with a PFS of more than 48 months compared with 19 months (p = 0.008). Considering all the results, our study suggested the need to integrate both the CD markers and copy number alterations to evaluate the prognosis of newly diagnosed multiple myeloma patients.
Collapse
Affiliation(s)
- Mihaiela L. Dragoș
- Biology Department, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700506 Iasi, Romania; (M.L.D.); (A.B.S.)
| | - Iuliu C. Ivanov
- Molecular Diagnosis Department, Regional Institute of Oncology, 700483 Iasi, Romania; (I.C.I.); (A.S.)
| | - Mihaela Mențel
- Center for Fundamental Research and Experimental Development in Translation Medicine—TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania; (M.M.); (I.C.V.-T.)
- Immunophenotyping Department, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Irina C. Văcărean-Trandafir
- Center for Fundamental Research and Experimental Development in Translation Medicine—TRANSCEND, Regional Institute of Oncology, 700483 Iasi, Romania; (M.M.); (I.C.V.-T.)
| | - Adriana Sireteanu
- Molecular Diagnosis Department, Regional Institute of Oncology, 700483 Iasi, Romania; (I.C.I.); (A.S.)
| | - Amalia A. Titianu
- Department of Hematology, Regional Institute of Oncology, 700483 Iasi, Romania; (A.A.T.); (A.S.D.)
- Department of Hematology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Angela S. Dăscălescu
- Department of Hematology, Regional Institute of Oncology, 700483 Iasi, Romania; (A.A.T.); (A.S.D.)
- Department of Hematology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Alexandru B. Stache
- Biology Department, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700506 Iasi, Romania; (M.L.D.); (A.B.S.)
- Molecular Diagnosis Department, Regional Institute of Oncology, 700483 Iasi, Romania; (I.C.I.); (A.S.)
| | - Daniela Jitaru
- Biology Department, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700506 Iasi, Romania; (M.L.D.); (A.B.S.)
| | - Dragoș L. Gorgan
- Biology Department, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, 700506 Iasi, Romania; (M.L.D.); (A.B.S.)
| |
Collapse
|
96
|
Davies FE, Pawlyn C, Usmani SZ, San-Miguel JF, Einsele H, Boyle EM, Corre J, Auclair D, Cho HJ, Lonial S, Sonneveld P, Stewart AK, Bergsagel PL, Kaiser MF, Weisel K, Keats JJ, Mikhael JR, Morgan KE, Ghobrial IM, Orlowski RZ, Landgren CO, Gay F, Caers J, Chng WJ, Chari A, Walker BA, Kumar SK, Costa LJ, Anderson KC, Morgan GJ. Perspectives on the Risk-Stratified Treatment of Multiple Myeloma. Blood Cancer Discov 2022; 3:273-284. [PMID: 35653112 PMCID: PMC9894570 DOI: 10.1158/2643-3230.bcd-21-0205] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The multiple myeloma treatment landscape has changed dramatically. This change, paralleled by an increase in scientific knowledge, has resulted in significant improvement in survival. However, heterogeneity remains in clinical outcomes, with a proportion of patients not benefiting from current approaches and continuing to have a poor prognosis. A significant proportion of the variability in outcome can be predicted on the basis of clinical and biochemical parameters and tumor-acquired genetic variants, allowing for risk stratification and a more personalized approach to therapy. This article discusses the principles that can enable the rational and effective development of therapeutic approaches for high-risk multiple myeloma.
Collapse
Affiliation(s)
| | - Charlotte Pawlyn
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, Department of Haematology, London, United Kingdom
| | - Saad Z. Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Jill Corre
- Unité de Génomique du Myélome, Institut Universitaire du Cancer, Toulouse France. Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Daniel Auclair
- The Multiple Myeloma Research Foundation, Norwalk, Connecticut
| | - Hearn Jay Cho
- The Multiple Myeloma Research Foundation, Norwalk, Connecticut
- Multiple Myeloma Center of Excellence, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Pieter Sonneveld
- Erasmus MC Cancer Institute, Department of Hematology, Rotterdam, the Netherlands
| | - A. Keith Stewart
- University Health Network and the Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Martin F. Kaiser
- The Royal Marsden Hospital, Department of Haematology, London, United Kingdom
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonathan J. Keats
- Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, Arizona
| | - Joseph R. Mikhael
- Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, Arizona
| | | | - Irene M. Ghobrial
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Robert Z. Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - C. Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Francesca Gay
- Division of Hematology, University of Torino, Torino, Italy
| | - Joseph Caers
- Department of Hematology, Centre Hospitalier Universitaire (CHU) de Liège, Liège, Belgium
| | - Wee Joo Chng
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University, Indianapolis, Indiana
- Department of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ajai Chari
- Multiple Myeloma Center of Excellence, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Brian A. Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University, Indianapolis, Indiana
| | - Shaji K. Kumar
- Department of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Luciano J. Costa
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kenneth C. Anderson
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
97
|
Sudha P, Ahsan A, Ashby C, Kausar T, Khera A, Kazeroun MH, Hsu CC, Wang L, Fitzsimons E, Salminen O, Blaney P, Czader M, Williams J, Abu Zaid MI, Ansari-Pour N, Yong KL, van Rhee F, Pierceall WE, Morgan GJ, Flynt E, Gooding S, Abonour R, Ramasamy K, Thakurta A, Walker BA. Myeloma Genome Project Panel is a Comprehensive Targeted Genomics Panel for Molecular Profiling of Patients with Multiple Myeloma. Clin Cancer Res 2022; 28:2854-2864. [PMID: 35522533 PMCID: PMC9250632 DOI: 10.1158/1078-0432.ccr-21-3695] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/11/2022] [Accepted: 05/03/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE We designed a comprehensive multiple myeloma targeted sequencing panel to identify common genomic abnormalities in a single assay and validated it against known standards. EXPERIMENTAL DESIGN The panel comprised 228 genes/exons for mutations, 6 regions for translocations, and 56 regions for copy number abnormalities (CNA). Toward panel validation, targeted sequencing was conducted on 233 patient samples and further validated using clinical FISH (translocations), multiplex ligation probe analysis (MLPA; CNAs), whole-genome sequencing (WGS; CNAs, mutations, translocations), or droplet digital PCR (ddPCR) of known standards (mutations). RESULTS Canonical immunoglobulin heavy chain translocations were detected in 43.2% of patients by sequencing, and aligned with FISH except for 1 patient. CNAs determined by sequencing and MLPA for 22 regions were comparable in 103 samples and concordance between platforms was R2 = 0.969. Variant allele frequency (VAF) for 74 mutations were compared between sequencing and ddPCR with concordance of R2 = 0.9849. CONCLUSIONS In summary, we have developed a targeted sequencing panel that is as robust or superior to FISH and WGS. This molecular panel is cost-effective, comprehensive, clinically actionable, and can be routinely deployed to assist risk stratification at diagnosis or posttreatment to guide sequencing of therapies.
Collapse
Affiliation(s)
- Parvathi Sudha
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Aarif Ahsan
- Translational Medicine, Bristol Myers Squibb, Summit, New Jersey
| | - Cody Ashby
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Tasneem Kausar
- Translational Medicine, Bristol Myers Squibb, Summit, New Jersey
| | - Akhil Khera
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Mohammad H. Kazeroun
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Chih-Chao Hsu
- Translational Medicine, Bristol Myers Squibb, Summit, New Jersey
| | - Lin Wang
- Department of Pathology and Laboratory Research, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | | | - Outi Salminen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Patrick Blaney
- Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Magdalena Czader
- Department of Pathology and Laboratory Research, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Jonathan Williams
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Mohammad I. Abu Zaid
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Naser Ansari-Pour
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Kwee L. Yong
- Cancer Institute, University College London, London, United Kingdom
| | - Frits van Rhee
- Myeloma Center, Winthrop P. Rockefeller Cancer institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Gareth J. Morgan
- Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, New Jersey
| | - Sarah Gooding
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Oxford Center for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - Rafat Abonour
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| | - Karthik Ramasamy
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford Center for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
- Radcliffe Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Anjan Thakurta
- Translational Medicine, Bristol Myers Squibb, Summit, New Jersey
- Oxford Center for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
- Radcliffe Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Brian A. Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana
| |
Collapse
|
98
|
Kumar S, Baizer L, Callander NS, Giralt SA, Hillengass J, Freidlin B, Hoering A, Richardson PG, Schwartz EI, Reiman A, Lentzsch S, McCarthy PL, Jagannath S, Yee AJ, Little RF, Raje NS. Gaps and opportunities in the treatment of relapsed-refractory multiple myeloma: Consensus recommendations of the NCI Multiple Myeloma Steering Committee. Blood Cancer J 2022; 12:98. [PMID: 35768410 PMCID: PMC9243011 DOI: 10.1038/s41408-022-00695-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 01/05/2023] Open
Abstract
A wide variety of new therapeutic options for Multiple Myeloma (MM) have recently become available, extending progression-free and overall survival for patients in meaningful ways. However, these treatments are not curative, and patients eventually relapse, necessitating decisions on the appropriate choice of treatment(s) for the next phase of the disease. Additionally, an important subset of MM patients will prove to be refractory to the majority of the available treatments, requiring selection of effective therapies from the remaining options. Immunomodulatory agents (IMiDs), proteasome inhibitors, monoclonal antibodies, and alkylating agents are the major classes of MM therapies, with several options in each class. Patients who are refractory to one agent in a class may be responsive to a related compound or to a drug from a different class. However, rules for selection of alternative treatments in these situations are somewhat empirical and later phase clinical trials to inform those choices are ongoing. To address these issues the NCI Multiple Myeloma Steering Committee formed a relapsed/refractory working group to review optimal treatment choices, timing, and sequencing and provide recommendations. Additional issues considered include the role of salvage autologous stem cell transplantation, risk stratification, targeted approaches for genetic subsets of MM, appropriate clinical trial endpoints, and promising investigational agents. This report summarizes the deliberations of the working group and suggests potential avenues of research to improve the precision, timing, and durability of treatments for Myeloma.
Collapse
Affiliation(s)
- Shaji Kumar
- Hematologic Malignancies, Mayo Clinic College of Medicine and Science, Rochester, USA
| | - Lawrence Baizer
- Division of Lung Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Natalie S Callander
- Myeloma Clinical Program, University of Wisconsin Carbone Cancer Center, Madison, USA
| | - Sergio A Giralt
- Division of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, Madison, USA
| | - Jens Hillengass
- Oncology and Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Boris Freidlin
- Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Antje Hoering
- Cancer Research and Biostatistics and University of Washington School of Public Health, Seattle, USA
| | - Paul G Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, USA
| | - Elena I Schwartz
- Coordinating Center for Clinical Trials, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anthony Reiman
- University of New Brunswick, Department of Medicine, Dalhousie University Department of Oncology, Saint John Regional Hospital, Fredericton, Canada
| | - Suzanne Lentzsch
- Multiple Myeloma and Amyloidosis Service, Department of Medicine, Columbia University Medical Center, New York, USA
| | - Philip L McCarthy
- Department of Medicine, Oncology and Internal Medicine, Transplant & Cellular Therapy Center, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Sundar Jagannath
- Division of Hematology and Medical Oncology, Mount Sinai School of Medicine, Center of Excellence for Multiple Myeloma, New York, USA
| | - Andrew J Yee
- Department of Medicine, Harvard Medical School, Multiple Myeloma Program, Medical Oncology, Massachusetts General Hospital, Boston, USA
| | - Richard F Little
- Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Noopur S Raje
- Department of Medicine, Harvard Medical School, Multiple Myeloma Program, Medical Oncology, Massachusetts General Hospital, Boston, USA
| |
Collapse
|
99
|
Kastritis E, Migkou M, Dalampira D, Gavriatopoulou M, Fotiou D, Roussou M, Kanellias N, Ntanasis-Stathopoulos I, Malandrakis P, Theodorakakou F, Sevastoudi A, Eleutherakis-Papaiakovou E, Triantafyllou T, Terpos E, Katodritou E, Dimopoulos MA. Chromosome 1q21 Aberrations Identify Ultra High-Risk Myeloma with Prognostic and Clinical Implications. Am J Hematol 2022; 97:1142-1149. [PMID: 35731917 DOI: 10.1002/ajh.26639] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/02/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022]
Abstract
Numerical abnormalities of chromosome 1q (+1q21) are common in patients with newly diagnosed multiple myeloma (MM) but their prognostic impact remains a matter of debate. In addition, the impact of the number of copies of 1q21 is not known. We analyzed 912 consecutive patients with symptomatic MM to evaluate the prognostic implications of +1q21 and of their copy number variations, as assessed by FISH. At the time of initial diagnosis 249 (27.3%) patients had +1q21, of which 150 (16.4%) had 3 copies and 99 (10.9%) had 4 or more copies. Presence of +1q21 was associated with advanced ISS stage (p=0.003), concurrent presence of other cytogenetics aberrations and advanced R-ISS stage (p<0.001). Patients with +1q21 had inferior PFS (median 34 vs 20 months, p<0.001) and OS (median 75 vs 44 months, p<0.001) but the copy number of 1q21 had no additional prognostic impact. In multivariate analysis, adjusting for R-ISS, age, treatment and HDM, +1q21 remained an independent prognostic factor both for PFS (p<0.001) and OS (p=0.008). The detrimental prognostic effect of +1q21 was more profound in R-ISS-3 patients, identifying a subgroup with OS of just 16 months (vs 46 for R-ISS-3 without +1q21, p<0.001). We further validated our findings in an independent cohort of 272 patients. In conclusion, presence of +1q21 is associated with more advanced disease, inferior PFS and OS but especially patients with R-ISS-3 disease and +1q21 have a very poor outcome comprising an ultra-high-risk group. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Magdalini Migkou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimitra Dalampira
- Department of Hematology, Theageneion Cancer Hospital, Thessaloniki, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Despina Fotiou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Maria Roussou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nikolaos Kanellias
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Foteini Theodorakakou
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | | | | | - Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Eirini Katodritou
- Department of Hematology, Theageneion Cancer Hospital, Thessaloniki, Greece
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
100
|
Bustoros M, Anand S, Sklavenitis-Pistofidis R, Redd R, Boyle EM, Zhitomirsky B, Dunford AJ, Tai YT, Chavda SJ, Boehner C, Neuse CJ, Rahmat M, Dutta A, Casneuf T, Verona R, Kastritis E, Trippa L, Stewart C, Walker BA, Davies FE, Dimopoulos MA, Bergsagel PL, Yong K, Morgan GJ, Aguet F, Getz G, Ghobrial IM. Genetic subtypes of smoldering multiple myeloma are associated with distinct pathogenic phenotypes and clinical outcomes. Nat Commun 2022; 13:3449. [PMID: 35705541 PMCID: PMC9200804 DOI: 10.1038/s41467-022-30694-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/13/2022] [Indexed: 12/12/2022] Open
Abstract
Smoldering multiple myeloma (SMM) is a precursor condition of multiple myeloma (MM) with significant heterogeneity in disease progression. Existing clinical models of progression risk do not fully capture this heterogeneity. Here we integrate 42 genetic alterations from 214 SMM patients using unsupervised binary matrix factorization (BMF) clustering and identify six distinct genetic subtypes. These subtypes are differentially associated with established MM-related RNA signatures, oncogenic and immune transcriptional profiles, and evolving clinical biomarkers. Three genetic subtypes are associated with increased risk of progression to active MM in both the primary and validation cohorts, indicating they can be used to better predict high and low-risk patients within the currently used clinical risk stratification models.
Collapse
Affiliation(s)
- Mark Bustoros
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
- Division of Hematology & Medical Oncology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Shankara Anand
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | | | - Robert Redd
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Eileen M Boyle
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | | | - Yu-Tzu Tai
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
| | - Selina J Chavda
- Division of Hematology, University College London, London, UK
| | - Cody Boehner
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
| | - Carl Jannes Neuse
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
- University of Münster Medical School, Münster, Germany
| | - Mahshid Rahmat
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
| | - Ankit Dutta
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
| | | | - Raluca Verona
- Janssen Research and Development, Spring House, PA, USA
| | - Efstathis Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Lorenzo Trippa
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chip Stewart
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Brian A Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Faith E Davies
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | | | - Kwee Yong
- Division of Hematology, University College London, London, UK
| | - Gareth J Morgan
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | | | - Gad Getz
- Broad Institute of MIT & Harvard, Cambridge, MA, USA.
- Department of Pathology, Massachusetts General Hospital Cancer Center, Boston, MA, USA.
| | - Irene M Ghobrial
- Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA.
- Broad Institute of MIT & Harvard, Cambridge, MA, USA.
| |
Collapse
|