1
|
Samur MK, Aktas Samur A, Shah P, Park JS, Fulciniti M, Shammas M, Corre J, Anderson KC, Parmigiani G, Avet-Loiseau H, Munshi NC. Development of hyperdiploidy starts at an early age and takes a decade to complete. Blood 2025; 145:520-525. [PMID: 39571146 DOI: 10.1182/blood.2024025250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 10/15/2024] [Indexed: 01/31/2025] Open
Abstract
ABSTRACT Nearly half of patients with multiple myeloma (MM) have hyperdiploidy (HMM) at diagnosis. Although HMM occurs early, the mutational processes before and after hyperdiploidy are still unclear. Here, we used 72 whole-genome sequencing samples from patients with HMM and identified pre- and post-HMM mutations to define the chronology of the development of hyperdiploidy. An MM cell accumulated a median of 0.56 mutations per megabase before HMM, and for every clonal pre-HMM mutation, 1.21 mutations per megabase accumulated after HMM. This analysis using mutations before and after hyperdiploidy shows that hyperdiploidy happens after somatic hypermutation. Prehyperdiploidy mutations are activation-induced cytidine deaminase and age/clock-like signature driven, whereas posthyperdiploidy mutations are from DNA damage and APOBEC. Interestingly, the first hyperdiploidy event occurred within the first 3 decades of life and took a decade to complete. Copy number changes affecting chromosomes 15 and 19 occurred first. Finally, mutations before initiating event affected chromosomes at different rates, whereas post-initiating event mutational processes affect each chromosome equally.
Collapse
Affiliation(s)
- Mehmet K Samur
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Anil Aktas Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Parth Shah
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Department of Hematology, Dartmouth Cancer Center, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire
| | - Joseph S Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Masood Shammas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jill Corre
- Unité de Genomique du Myélome, University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Giovanni Parmigiani
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Hervé Avet-Loiseau
- Unité de Genomique du Myélome, University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Nikhil C Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
- Veterans Affairs Boston Healthcare System, Boston, MA
| |
Collapse
|
2
|
Monchusi B, Dube P, Takundwa MM, Kenmogne VL, Thimiri Govinda Raj DB. Advances in CRISPR-Cas systems for blood cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 208:261-284. [PMID: 39266186 DOI: 10.1016/bs.pmbts.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
CRISPR-Cas systems have revolutionised precision medicine by enabling personalised treatments tailored to an individual's genetic profile. Various CRISPR technologies have been developed to target specific disease-causing genes in blood cancers, and some have advanced to clinical trials. Although some studies have explored the in vivo applications of CRISPR-Cas systems, several challenges continue to impede their widespread use. Furthermore, CRISPR-Cas technology has shown promise in improving the response of immunotherapies to blood cancers. The emergence of CAR-T cell therapy has shown considerable success in the targeting and correcting of disease-causing genes in blood cancers. Despite the promising potential of CRISPR-Cas in the treatment of blood cancers, issues related to safety, ethics, and regulatory approval remain significant hurdles. This comprehensive review highlights the transformative potential of CRISPR-Cas technology to revolutionise blood cancer therapy.
Collapse
Affiliation(s)
- Bernice Monchusi
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Phumuzile Dube
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Mutsa Monica Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Vanelle Larissa Kenmogne
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa; Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - Deepak Balaji Thimiri Govinda Raj
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
| |
Collapse
|
3
|
Li JR, Arsang-Jang S, Cheng Y, Sun F, D'Souza A, Dhakal B, Hari P, Huang Q, Auer P, Li Y, Urrutia R, Zhan F, Shaughnessy JD, Janz S, Dong J, Cheng C. Enhancing prognostic power in multiple myeloma using a plasma cell signature derived from single-cell RNA sequencing. Blood Cancer J 2024; 14:38. [PMID: 38443358 PMCID: PMC10915134 DOI: 10.1038/s41408-024-01024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024] Open
Abstract
Multiple myeloma (MM) is a heterogenous plasma cell malignancy, for which the established prognostic models exhibit limitations in capturing the full spectrum of outcome variability. Leveraging single-cell RNA-sequencing data, we developed a novel plasma cell gene signature. We evaluated and validated the associations of the resulting plasma cell malignancy (PBM) score with disease state, progression and clinical outcomes using data from five independent myeloma studies consisting of 2115 samples (1978 MM, 65 monoclonal gammopathy of undetermined significance, 35 smoldering MM, and 37 healthy controls). Overall, a higher PBM score was significantly associated with a more advanced stage within the spectrum of plasma cell dyscrasias (all p < 0.05) and a shorter overall survival in MM (hazard ratio, HR = 1.72; p < 0.001). Notably, the prognostic effect of the PBM score was independent of the International Staging System (ISS) and Revised ISS (R-ISS). The downstream analysis further linked higher PBM scores with the presence of cytogenetic abnormalities, TP53 mutations, and compositional changes in the myeloma tumor immune microenvironment. Our integrated analyses suggest the PBM score may provide an opportunity for refining risk stratification and guide decisions on therapeutic approaches to MM.
Collapse
Affiliation(s)
- Jian-Rong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
| | - Shahram Arsang-Jang
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yan Cheng
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Fumou Sun
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Anita D'Souza
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Binod Dhakal
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Parameswaran Hari
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Quillan Huang
- Department of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Paul Auer
- Division of Biostatistics, Institute for Health & Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Raul Urrutia
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John D Shaughnessy
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Siegfried Janz
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jing Dong
- Division of Hematology Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
4
|
Terragna C, Poletti A, Solli V, Martello M, Zamagni E, Pantani L, Borsi E, Vigliotta I, Mazzocchetti G, Armuzzi S, Taurisano B, Testoni N, Marzocchi G, Kanapari A, Pistis I, Tacchetti P, Mancuso K, Rocchi S, Rizzello I, Cavo M. Multi-dimensional scaling techniques unveiled gain1q&loss13q co-occurrence in Multiple Myeloma patients with specific genomic, transcriptional and adverse clinical features. Nat Commun 2024; 15:1551. [PMID: 38378709 PMCID: PMC10879136 DOI: 10.1038/s41467-024-45000-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
The complexity of Multiple Myeloma (MM) is driven by several genomic aberrations, interacting with disease-related and/or -unrelated factors and conditioning patients' clinical outcome. Patient's prognosis is hardly predictable, as commonly employed MM risk models do not precisely partition high- from low-risk patients, preventing the reliable recognition of early relapsing/refractory patients. By a dimensionality reduction approach, here we dissect the genomic landscape of a large cohort of newly diagnosed MM patients, modelling all the possible interactions between any MM chromosomal alterations. We highlight the presence of a distinguished cluster of patients in the low-dimensionality space, with unfavorable clinical behavior, whose biology was driven by the co-occurrence of chromosomes 1q CN gain and 13 CN loss. Presence or absence of these alterations define MM patients overexpressing either CCND2 or CCND1, fostering the implementation of biology-based patients' classification models to describe the different MM clinical behaviors.
Collapse
Affiliation(s)
- Carolina Terragna
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy.
| | - Andrea Poletti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Vincenza Solli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Marina Martello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Lucia Pantani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Enrica Borsi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Ilaria Vigliotta
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Gaia Mazzocchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Silvia Armuzzi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Barbara Taurisano
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Nicoletta Testoni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Giulia Marzocchi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Ajsi Kanapari
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Ignazia Pistis
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Katia Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Serena Rocchi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Ilaria Rizzello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna-Istituto di Ematologia "Seràgnoli", Bologna, Italy
- DIMEC-Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Buenache N, Sánchez-delaCruz A, Cuenca I, Giménez A, Moreno L, Martínez-López J, Rosa-Rosa JM. Identification of Immunoglobulin Gene Rearrangement Biomarkers in Multiple Myeloma through cfDNA-Based Liquid Biopsy Using tchDNA-Seq. Cancers (Basel) 2023; 15:cancers15112911. [PMID: 37296872 DOI: 10.3390/cancers15112911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the clonal proliferation of pathogenic CD138+ plasma cells (PPCs) in bone marrow (BM). Recent years have seen a significant increase in the treatment options for MM; however, most patients who achieve complete the response ultimately relapse. The earlier detection of tumor-related clonal DNA would thus be very beneficial for patients with MM and would enable timely therapeutic interventions to improve outcomes. Liquid biopsy of "cell-free DNA" (cfDNA) as a minimally invasive approach might be more effective than BM aspiration not only for the diagnosis but also for the detection of early recurrence. Most studies thus far have addressed the comparative quantification of patient-specific biomarkers in cfDNA with PPCs and BM samples, which have shown good correlations. However, there are limitations to this approach, such as the difficulty in obtaining enough circulating free tumor DNA to achieve sufficient sensitivity for the assessment of minimal residual disease. Herein, we summarize current data on methodologies to characterize MM, and we present evidence that targeted capture hybridization DNA sequencing (tchDNA-Seq) can provide robust biomarkers in cfDNA, including immunoglobulin (IG) rearrangements. We also show that detection can be improved by prior purification of the cfDNA. Overall, liquid biopsies of cfDNA to monitor IG rearrangements have the potential to provide important diagnostic, prognostic, and predictive information in patients with MM.
Collapse
Affiliation(s)
- Natalia Buenache
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Andrea Sánchez-delaCruz
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Isabel Cuenca
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Alicia Giménez
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Laura Moreno
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
| | - Joaquín Martínez-López
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
- Department of Translational Haematology, Haematology Service, Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Medicine, Faculty of Medicine, Complutense University, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28034 Madrid, Spain
| | - Juan Manuel Rosa-Rosa
- Department of Translational Haematology, Research Institute Hospital 12 de Octubre (i+12) Haematological Tumors Clinical Research Unit H12O-CNIO, 28041 Madrid, Spain
- Spanish National Cancer Research Center (CNIO), 28034 Madrid, Spain
| |
Collapse
|
8
|
Li W, Zhang B, Cao W, Zhang W, Li T, Liu L, Xu L, Gao F, Wang Y, Wang F, Xing H, Jiang Z, Shi J, Bian Z, Song Y. Identification of potential resistance mechanisms and therapeutic targets for the relapse of BCMA CAR-T therapy in relapsed/refractory multiple myeloma through single-cell sequencing. Exp Hematol Oncol 2023; 12:44. [PMID: 37158921 PMCID: PMC10165782 DOI: 10.1186/s40164-023-00402-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND BCMA CAR-T is highly effective for relapsed/refractory multiple myeloma(R/R-MM) and significantly improves the survival of patients. However, the short remission time and high relapse rate of MM patients treated with BCMA CAR-T remain bottlenecks that limit long-term survival. The immune microenvironment of the bone marrow (BM) in R/R-MM may be responsible for this. The present study aims to present an in-depth analysis of resistant mechanisms and to explore potential novel therapeutic targets for relapse of BCMA CAR-T treatment via single-cell RNA sequencing (scRNA-seq) of BM plasma cells and immune cells. METHODS This study used 10X Genomic scRNA-seq to identify cell populations in R/R-MM CD45+ BM cells before BCMA CAR-T treatment and relapse after BCMA CAR-T treatment. Cell Ranger pipeline and CellChat were used to perform detailed analysis. RESULTS We compared the heterogeneity of CD45+ BM cells before BCMA CAR-T treatment and relapse after BCMA CAR-T treatment. We found that the proportion of monocytes/macrophages increased, while the percentage of T cells decreased at relapse after BCMA CAR-T treatment. We then reclustered and analyzed the alterations in plasma cells, T cells, NK cells, DCs, neutrophils, and monocytes/macrophages in the BM microenvironment before BCMA CAR-T treatment and relapse after BCMA CAR-T treatment. We show here that the percentage of BCMA positive plasma cells increased at relapse after BCMA CAR-T cell therapy. Other targets such as CD38, CD24, SLAMF7, CD138, and GPRC5D were also found to be expressed in plasma cells of the R/R-MM patient at relapse after BCMA CAR-T cell therapy. Furthermore, exhausted T cells, TIGIT+NK cells, interferon-responsive DCs, and interferon-responsive neutrophils, increased in the R/R-MM patient at relapse after BCMA CAR-T cell treatment. Significantly, the proportion of IL1βhi Mφ, S100A9hi Mφ, interferon-responsive Mφ, CD16hi Mφ, MARCO hi Mφ, and S100A11hi Mφ significantly increased in the R/R-MM patient at relapse after BCMA CAR-T cell therapy. Cell-cell communication analysis indicated that monocytes/macrophages, especially the MIF and APRIL signaling pathway are key players in R/R-MM patient at relapse after BCMA CAR-T cell therapy. CONCLUSION Taken together, our data extend the understanding of intrinsic and extrinsic relapse of BCMA CAR-T treatment in R/R-MM patient and the potential mechanisms involved in the alterations of antigens and the induced immunosuppressive microenvironment, which may provide a basis for the optimization of BCMA CAR-T strategies. Further studies should be performed to confirm these findings.
Collapse
Affiliation(s)
- Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Binglei Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Wenli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Lina Liu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - LinPing Xu
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Fengcai Gao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Yanmei Wang
- Department of Hematology, Zhengzhou People's Hospital, Zhengzhou, 450003, Henan, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Haizhou Xing
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Jianxiang Shi
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China.
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
9
|
Sun K, Jin L, Karolová J, Vorwerk J, Hailfinger S, Opalka B, Zapukhlyak M, Lenz G, Khandanpour C. Combination Treatment Targeting mTOR and MAPK Pathways Has Synergistic Activity in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15082373. [PMID: 37190302 DOI: 10.3390/cancers15082373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Multiple myeloma (MM) is an incurable, malignant B cell disorder characterized by frequent relapses and a poor prognosis. Thus, new therapeutic approaches are warranted. The phosphatidylinositol-3-kinase (PI3K) pathway plays a key role in many critical cellular processes, including cell proliferation and survival. Activated PI3K/AKT (protein kinases B)/mTOR (mammalian target of rapamycin) signaling has been identified in MM primary patient samples and cell lines. In this study, the efficacy of PI3K and mTOR inhibitors in various MM cell lines representing three different prognostic subtypes was tested. Whereas MM cell lines were rather resistant to PI3K inhibition, treatment with the mTOR inhibitor temsirolimus decreases the phosphorylation of key molecules in the PI3K pathway in MM cell lines, leading to G0/G1 cell cycle arrest and thus reduced proliferation. Strikingly, the efficacy of temsirolimus was amplified by combining the treatment with the Mitogen-activated protein kinase kinase (MEK) inhibitor trametinib. Our findings provide a scientific rationale for the simultaneous inhibition of mTOR and MEK as a novel strategy for the treatment of MM.
Collapse
Affiliation(s)
- Kaiyan Sun
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Ling Jin
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Jana Karolová
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, 12108 Prague, Czech Republic
| | - Jan Vorwerk
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Stephan Hailfinger
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Bertram Opalka
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center (WTZ), University Hospital Essen, 45147 Essen, Germany
| | - Myroslav Zapukhlyak
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, Hematology, Hemostaseology, Oncology and Pneumology, University Hospital Münster, 48149 Münster, Germany
- Department of Hematology and Oncology, University Hospital Schleswig-Holstein and University of Lübeck, 23538 Lübeck, Germany
| |
Collapse
|
10
|
Aarabi M, Yoest JM, Farah R, Rajkovic A, Swerdlow SH, Yatsenko SA. A Novel Integrated Approach for Cytogenomic Evaluation of Plasma Cell Neoplasms. J Mol Diagn 2022; 24:1067-1078. [PMID: 35940519 DOI: 10.1016/j.jmoldx.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/24/2022] [Accepted: 07/19/2022] [Indexed: 12/29/2022] Open
Abstract
Plasma cell neoplasm (PCN) is associated with characteristic chromosomal aberrations of diagnostic and prognostic significance. The presence of a small percentage of neoplastic cells is a drawback in the application of karyotyping and fluorescence in situ hybridization for the evaluation of bone marrow aspirate. The analysis of samples enriched for CD138+ cells has improved the detection rate. However, fluorescence in situ hybridization requires several probes and may not be completed due to a limited number of isolated cells. To address the issues experienced with the conventional approach, a novel integrated protocol that consists of whole-genome amplification of DNA isolated from CD138+ cells, followed by microarray as well as one fluorescence in situ hybridization assay for balanced IGH gene rearrangements, has been developed. In the present study in a cohort of 56 patients with clinical suspicion for PCN, compared to conventional cytogenetic analysis, this approach provided higher yield in the detection of PCN-related abnormalities, irrespective of the initial percentage of plasma cells. Whole-genome profiling uncovered recurrent chromosomal abnormalities of prognostic value, including unbalanced alterations within the MYC locus, 16q loss, and hypodiploidy, that were not otherwise detectable by conventional methods. The proposed approach is cost-efficient and provides a superior detection rate, required for proper risk stratification and differential diagnosis of PCN regardless of initial plasma cell percentage.
Collapse
Affiliation(s)
- Mahmoud Aarabi
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer M Yoest
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rafic Farah
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Aleksandar Rajkovic
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Pathology, University of California-San Francisco, San Francisco, California; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California-San Francisco, San Francisco, California; Institute of Human Genetics, University of California-San Francisco, San Francisco, California
| | - Steven H Swerdlow
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Svetlana A Yatsenko
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Diamantidis MD, Papadaki S, Hatjiharissi E. Exploring the current molecular landscape and management of multiple myeloma patients with the t(11;14) translocation. Front Oncol 2022; 12:934008. [PMID: 35982976 PMCID: PMC9379277 DOI: 10.3389/fonc.2022.934008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is a genetically complex disease. The key myeloma-initiating genetic events are hyperdiploidy and translocations involving the immunoglobulin heavy chain (IgH) enhancer on chromosome 14, which leads to the activation of oncogenes (e.g., CCND1, CCND3, MAF, and MMSET). The t(11;14) translocation is the most common in MM (15%–20%) and results in cyclin D1 (CCND1) upregulation, which leads to kinase activation and tumor cell proliferation. Notably, t(11;14) occurs at a higher rate in patients with plasma cell leukemia (40%) and light chain amyloidosis (50%). Patients with myeloma who harbor the t(11;14) translocation have high levels of the anti-apoptotic protein B-cell lymphoma 2 (BCL2). Multiple studies demonstrated that the presence of t(11;14) was predictive of BCL2 dependency, suggesting that BCL2 could be a target in this subtype of myeloma. Venetoclax, an oral BCL2 inhibitor, has shown remarkable activity in treating relapsed/refractory MM patients with t(11;14) and BCL2 overexpression, either as monotherapy or in combination with other anti-myeloma agents. In this review, we describe the molecular defects associated with the t(11;14), bring into question the standard cytogenetic risk of myeloma patients harboring t(11;14), summarize current efficacy and safety data of targeted venetoclax-based therapies, and discuss the future of individualized or precision medicine for this unique myeloma subgroup, which will guide optimal treatment.
Collapse
Affiliation(s)
- Michael D. Diamantidis
- Thalassemia and Sickle Cell Disease Unit, Department of Hematology, General Hospital of Larissa, Larissa, Greece
| | - Sofia Papadaki
- Division of Hematology, First Department of Internal Medicine, AHEPA General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evdoxia Hatjiharissi
- Division of Hematology, First Department of Internal Medicine, AHEPA General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
- *Correspondence: Evdoxia Hatjiharissi,
| |
Collapse
|
14
|
Amundarain A, Valcárcel LV, Ordoñez R, Garate L, Miranda E, Cendoya X, Carrasco‐Leon A, Calasanz MJ, Paiva B, Meydan C, Mason CE, Melnick A, Rodriguez‐Otero P, Martín‐Subero JI, San Miguel J, Planes FJ, Prósper F, Agirre X. Landscape and clinical significance of long noncoding RNAs involved in multiple myeloma expressed fusion transcripts. Am J Hematol 2022; 97:E113-E117. [PMID: 34961980 DOI: 10.1002/ajh.26450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/31/2022]
Affiliation(s)
- Ane Amundarain
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
| | - Luis V. Valcárcel
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Tecnun School of Engineering, Biomedical Engineering Center University of Navarra San Sebastian Spain
| | - Raquel Ordoñez
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
| | - Leire Garate
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- Hematology Department, Clínica Universidad de Navarra University of Navarra Pamplona Spain
| | - Estíbaliz Miranda
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
| | - Xabier Cendoya
- Tecnun School of Engineering, Biomedical Engineering Center University of Navarra San Sebastian Spain
| | - Arantxa Carrasco‐Leon
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
| | - María José Calasanz
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- CIMA LAB Diagnostics University of Navarra Pamplona Spain
| | - Bruno Paiva
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- Hematology Department, Clínica Universidad de Navarra University of Navarra Pamplona Spain
- CIMA LAB Diagnostics University of Navarra Pamplona Spain
- Flow Cytometry Core, CIMA University of Navarra Pamplona Spain
| | - Cem Meydan
- Division of Hematology/Oncology, Department of Medicine Weill Cornell Medical College New York New York USA
- Department of Physiology and Biophysics Weill Cornell Medicine New York New York USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine Weill Cornell Medicine New York New York USA
| | - Christopher E. Mason
- Division of Hematology/Oncology, Department of Medicine Weill Cornell Medical College New York New York USA
- Department of Physiology and Biophysics Weill Cornell Medicine New York New York USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine Weill Cornell Medicine New York New York USA
| | - Ari Melnick
- Division of Hematology/Oncology, Department of Medicine Weill Cornell Medical College New York New York USA
| | - Paula Rodriguez‐Otero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- Hematology Department, Clínica Universidad de Navarra University of Navarra Pamplona Spain
| | - José I. Martín‐Subero
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- Hospital Clínic de Barcelona and Departament de Fonaments Clínics, Facultat de Medicina Universitat de Barcelona Barcelona Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer IDIBAPS Barcelona Spain
- Institució Catalana de Recerca i Estudis Avançats ICREA Barcelona Spain
| | - Jesús San Miguel
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- Hematology Department, Clínica Universidad de Navarra University of Navarra Pamplona Spain
| | - Francisco J. Planes
- Tecnun School of Engineering, Biomedical Engineering Center University of Navarra San Sebastian Spain
| | - Felipe Prósper
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
- Hematology Department, Clínica Universidad de Navarra University of Navarra Pamplona Spain
| | - Xabier Agirre
- Hemato‐Oncology Program, Center for Applied Medical Research (CIMA), IDISNA University of Navarra Pamplona Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) Pamplona Spain
| |
Collapse
|
15
|
Xu J, Wang Y, Wei Z, Zhuang J, Li J, Sun Y, Ren L, Wang Y, Li P, Gu S, Zhang Y, Jiang J, Chen C, Zhang Y, Liu P. Single-Cell Transcriptomes Combining with Consecutive Genomics Reveal Clonal Evolution and Gene Regulatory Networks in Relapsed and Refractory Multiple Myeloma. Front Cell Dev Biol 2022; 9:794144. [PMID: 35071234 PMCID: PMC8766805 DOI: 10.3389/fcell.2021.794144] [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: 10/13/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022] Open
Abstract
This study attempted to investigate how clonal structure evolves, along with potential regulatory networks, as a result of multiline therapies in relapsed/refractory multiple myeloma (RRMM). Eight whole exome sequencing (WES) and one single cell RNA sequencing (scRNA-seq) were performed in order to assess dynamic genomic changes in temporal consecutive samples of one RRMM patient from the time of diagnosis to death (about 37 months). The 63-year-old female patient who suffered from MM (P1) had disease progression (PD) nine times from July 2017 [newly diagnosed (ND)] to Aug 2020 (death), and the force to drive branching-pattern evolution of malignant PCs was found to be sustained. The mutant-allele tumor heterogeneity (MATH) and tumor mutation burden (TMB) initially exhibited a downward trend, which was then upward throughout the course of the disease. Various somatic single nucleotide variants (SNVs) that had disappeared after the previous treatment were observed to reappear in later stages. Chromosomal instability (CIN) and homologous recombination deficiency (HRD) scores were observed to be increased during periods of all progression, especially in the period of extramedullary plasmacytoma. Finally, in combination with WES and scRNA-seq of P1-PD9 (the nineth PD), the intro-heterogeneity and gene regulatory networks of MM cells were deciphered. As verified by the overall survival of MM patients in the MMRF CoMMpass and GSE24080 datasets, RUNX3 was identified as a potential driver for RRMM.
Collapse
Affiliation(s)
- Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Wei
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingli Zhuang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifeng Sun
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liang Ren
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yawen Wang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Panpan Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shiyang Gu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yian Zhang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jifeng Jiang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
YTHDF2 promotes multiple myeloma cell proliferation via STAT5A/MAP2K2/p-ERK axis. Oncogene 2022; 41:1482-1491. [PMID: 35075244 DOI: 10.1038/s41388-022-02191-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 12/21/2021] [Accepted: 01/12/2022] [Indexed: 12/27/2022]
Abstract
Multiple myeloma (MM) is still incurable partially due to lacking effective therapeutic targets. Aberrant N6-methyladenosine (m6A) RNA modification plays a vital role in many cancers, however few researches are executed in MM. We first screened the m6A-related genes in MM patient cohorts and correlated these genes with patient outcomes. We found that YTHDF2, a well-recognized m6A reader, was increased in MM patients and associated with poor outcomes. Decreased YTHDF2 expression hampered MM cell proliferation in vitro and in vivo, while enforced YTHDF2 expression reversed those effects. The analyses of m6A-RIP-seq and RIP-PCR indicated that STAT5A was the downstream target of YTHDF2, which was binding to the m6A modification site of STAT5A to promote its mRNA degradation. ChIP-seq and PCR assays revealed that STAT5A suppressed MM cell proliferation by occupying the transcription site of MAP2K2 to decrease ERK phosphorylation. In addition, we confirmed that YTHDF2 mediated the unphosphorylated form of STAT5A to inhibit the expression of MAP2K2/p-ERK. In conclusion, our study highlights that YTHDF2/STAT5A/MAP2K2/p-ERK axis plays a key role in MM proliferation and targeting YTHDF2 may be a promising therapeutic strategy.
Collapse
|
17
|
Hussen BM, Abdullah ST, Salihi A, Sabir DK, Sidiq KR, Rasul MF, Hidayat HJ, Ghafouri-Fard S, Taheri M, Jamali E. The emerging roles of NGS in clinical oncology and personalized medicine. Pathol Res Pract 2022; 230:153760. [PMID: 35033746 DOI: 10.1016/j.prp.2022.153760] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 02/07/2023]
Abstract
Next-generation sequencing (NGS) has been increasingly popular in genomics studies over the last decade, as new sequencing technology has been created and improved. Recently, NGS started to be used in clinical oncology to improve cancer therapy through diverse modalities ranging from finding novel and rare cancer mutations, discovering cancer mutation carriers to reaching specific therapeutic approaches known as personalized medicine (PM). PM has the potential to minimize medical expenses by shifting the current traditional medical approach of treating cancer and other diseases to an individualized preventive and predictive approach. Currently, NGS can speed up in the early diagnosis of diseases and discover pharmacogenetic markers that help in personalizing therapies. Despite the tremendous growth in our understanding of genetics, NGS holds the added advantage of providing more comprehensive picture of cancer landscape and uncovering cancer development pathways. In this review, we provided a complete overview of potential NGS applications in scientific and clinical oncology, with a particular emphasis on pharmacogenomics in the direction of precision medicine treatment options.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq; Center of Research and Strategic Studies, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Sara Tharwat Abdullah
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Abbas Salihi
- Center of Research and Strategic Studies, Lebanese French University, Kurdistan Region, Erbil, Iraq; Department of Biology, College of Science, Salahaddin University, Kurdistan Region, Erbil, Iraq
| | - Dana Khdr Sabir
- Department of Medical Laboratory Sciences, Charmo University, Kurdistan Region, Iraq
| | - Karzan R Sidiq
- Department of Biology, College of Education, University of Sulaimani, Sulaimani 334, Kurdistan, Iraq
| | - Mohammed Fatih Rasul
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Kurdistan Region, Erbil, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University, Kurdistan Region, Erbil, Iraq
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany; Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elena Jamali
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Black H, Glavey S. Gene expression profiling as a prognostic tool in multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:1008-1018. [PMID: 35582380 PMCID: PMC8992436 DOI: 10.20517/cdr.2021.83] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/12/2021] [Accepted: 10/27/2021] [Indexed: 11/12/2022]
Abstract
Multiple myeloma (MM) is an aggressive plasma cell malignancy with high degrees of variability in outcome, some patients experience long remissions, whilst others survive less than two years from diagnosis. Therapy refractoriness and relapse remain challenges in MM management, and there is a need for improved prognostication and targeted therapies to improve overall survival (OS). The past decade has seen a surge in gene expression profiling (GEP) studies which have elucidated the molecular landscape of MM and led to the identification of novel gene signatures that predict OS and outperform current clinical predictors. In this review, we discuss the limitations of current prognostic tools and the emerging role of GEP in diagnostics and in the development of personalised medicine approaches to combat drug resistance.
Collapse
Affiliation(s)
- Harmony Black
- Department of Haematology, Beaumont Hospital, Dublin D09 V2N0, Ireland
| | - Siobhan Glavey
- Department of Haematology, Beaumont Hospital, Dublin D09 V2N0, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, Dublin D02 YN77, Ireland
| |
Collapse
|
19
|
Puła A, Robak P, Mikulski D, Robak T. The Significance of mRNA in the Biology of Multiple Myeloma and Its Clinical Implications. Int J Mol Sci 2021; 22:12070. [PMID: 34769503 PMCID: PMC8584466 DOI: 10.3390/ijms222112070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a genetically complex disease that results from a multistep transformation of normal to malignant plasma cells in the bone marrow. However, the molecular mechanisms responsible for the initiation and heterogeneous evolution of MM remain largely unknown. A fundamental step needed to understand the oncogenesis of MM and its response to therapy is the identification of driver mutations. The introduction of gene expression profiling (GEP) in MM is an important step in elucidating the molecular heterogeneity of MM and its clinical relevance. Since some mutations in myeloma occur in non-coding regions, studies based on the analysis of mRNA provide more comprehensive information on the oncogenic pathways and mechanisms relevant to MM biology. In this review, we discuss the role of gene expression profiling in understanding the biology of multiple myeloma together with the clinical manifestation of the disease, as well as its impact on treatment decisions and future directions.
Collapse
Affiliation(s)
- Anna Puła
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland;
| | - Paweł Robak
- Department of Experimental Hematology, Medical University of Lodz, 93-510 Lodz, Poland;
| | - Damian Mikulski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland;
| |
Collapse
|
20
|
How I Treat High-risk Multiple Myeloma. Blood 2021; 139:2889-2903. [PMID: 34727187 DOI: 10.1182/blood.2020008733] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/26/2021] [Indexed: 11/20/2022] Open
Abstract
Survival of multiple myeloma (MM) has significantly improved over the last decade; however, a composed group of patients (15-20%), named high-risk (HR) MM, still experience reduced survival. Both tumor biology and suboptimal/absent responses to therapy may underlie HR definition and a clear uniform identification of risk factors is crucial for a proper management of these patients. In biologic-HRMM, MRD negativity attainment and sustain, inside and outside BM, should be the primary goal and therapy should be adapted in patients with frailty to reduce toxicity and improve quality of life. MM treatment has traditionally been tailored on age and more recently frailty or comorbidities, but very rarely on the biology of the disease, mainly because of the lack of a clear benefit derived from a specific drug/combination, inhomogeneity in HR definition and lack of data coming from prospective, properly designed clinical trials. Some attempts have been successfully made recently in this direction. In this review, we are discussing the current different definitions of HR and the need for a consensus, the results of available trials in HR patients and the way through risk-adapted treatment strategies. For this purpose, we are proposing several clinical cases of difficult-to-treat patients throughout different treatment phases.
Collapse
|
21
|
Cutler SD, Knopf P, Campbell CJV, Thoni A, El Hassan MA, Forward N, White D, Wagner J, Goudie M, Boudreau JE, Kennedy BE, Gujar S, Gaston D, Elnenaei MO. DMG26: A Targeted Sequencing Panel for Mutation Profiling to Address Gaps in the Prognostication of Multiple Myeloma. J Mol Diagn 2021; 23:1699-1714. [PMID: 34562616 DOI: 10.1016/j.jmoldx.2021.08.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/15/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022] Open
Abstract
Multiple myeloma presents with numerous primary genomic lesions that broadly dichotomize cases into hyperdiploidy or IgH translocated. Clinically, these large alterations are assessed by fluorescence in situ hybridization (FISH) for risk stratification at diagnosis. Secondary focal events, including indels and single-nucleotide variants, are also reported; however, their clinical correlates are poorly described, and FISH has insufficient resolution to assess many of them. In this study, we examined the exonic sequences of 26 genes reported to be mutated in >1% of patients with myeloma using a custom panel. We sequenced these exons to approximately 1000 times in a cohort of 76 patients from Atlantic Canada with detailed clinical correlates and in four multiple myeloma cell lines. Across the 76 patients, 255 mutations and 33 focal copy number variations were identified. High-severity mutations and mutations predicted by FATHMM-XF to be pathogenic identified patients with significantly reduced progression-free survival. These mutations were mutually exclusive from the Revised International Staging System high-risk FISH markers and were independent of all biochemical parameters of the Revised International Staging System. Applying our panel to patients classified by FISH to be standard risk successfully reclassified patients into high- and standard-risk groups. Furthermore, three patients in our cohort each had two high-risk markers; two of these patients developed plasma cell leukemia, a rare and severe clinical sequela of multiple myeloma.
Collapse
Affiliation(s)
- Samuel D Cutler
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Philipp Knopf
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Clinton J V Campbell
- Pathology & Molecular Medicine, Faculty of Health Sciences, McMaster University, Toronto, Ontario, Canada
| | - Andrea Thoni
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada; Pathology & Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | | | - Nicholas Forward
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Darrell White
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Julie Wagner
- Pathology & Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Marissa Goudie
- Pathology & Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Jeanette E Boudreau
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Barry E Kennedy
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Shashi Gujar
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Daniel Gaston
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada; Pathology & Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.
| | - Manal O Elnenaei
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada; Pathology & Laboratory Medicine, Nova Scotia Health, Halifax, Nova Scotia, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.
| |
Collapse
|
22
|
Gowin K, Skerget S, Keats JJ, Mikhael J, Cowan AJ. Plasma cell leukemia: A review of the molecular classification, diagnosis, and evidenced-based treatment. Leuk Res 2021; 111:106687. [PMID: 34425325 DOI: 10.1016/j.leukres.2021.106687] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/22/2021] [Accepted: 08/15/2021] [Indexed: 12/11/2022]
Abstract
Plasma cell leukemia is a rare and aggressive plasma cell dyscrasia associated with dismal outcomes. It may arise de novo, primary plasma cell leukemia, or evolve from an antecedent diagnosis of multiple myeloma, secondary plasma cell leukemia. Despite highly effective therapeutics, survival for plasma cell leukemia patients remains poor. Molecular knowledge of plasma cell leukemia has recently expanded with use of gene expression profiling and whole exome sequencing, lending new insights into prognosis and therapeutic development. In this review, we describe the molecular knowledge, clinical characteristics, evidenced-based therapeutic approaches and treatment outcomes of plasma cell leukemia.
Collapse
Affiliation(s)
- Krisstina Gowin
- University of Arizona, Department of Bone Marrow Transplant and Cellular Therapy, Tucson, AZ, United States.
| | - Sheri Skerget
- Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, United States
| | - Jonathan J Keats
- Translational Genomics Research Institute (TGen), Integrated Cancer Genomics Division, Phoenix, AZ, United States
| | - Joseph Mikhael
- Translational Genomics Research Institute (TGen), Applied Cancer Research and Drug Discovery Division, Phoenix, AZ, United States
| | - Andrew J Cowan
- University of Washington, Department of Hematology Oncology, Seattle, WA, United States
| |
Collapse
|
23
|
A Comprehensive Review of the Genomics of Multiple Myeloma: Evolutionary Trajectories, Gene Expression Profiling, and Emerging Therapeutics. Cells 2021; 10:cells10081961. [PMID: 34440730 PMCID: PMC8391934 DOI: 10.3390/cells10081961] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 12/16/2022] Open
Abstract
Multiple myeloma (MM) is a blood cancer characterized by the accumulation of malignant monoclonal plasma cells in the bone marrow. It develops through a series of premalignant plasma cell dyscrasia stages, most notable of which is the Monoclonal Gammopathy of Undetermined Significance (MGUS). Significant advances have been achieved in uncovering the genomic aberrancies underlying the pathogenesis of MGUS-MM. In this review, we discuss in-depth the genomic evolution of MM and focus on the prognostic implications of the accompanied molecular and cytogenetic aberrations. We also dive into the latest investigatory techniques used for the diagnoses and risk stratification of MM patients.
Collapse
|
24
|
Zanwar S, Kumar S. Disease heterogeneity, prognostication and the role of targeted therapy in multiple myeloma. Leuk Lymphoma 2021; 62:3087-3097. [PMID: 34304677 DOI: 10.1080/10428194.2021.1957875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma cell malignancy with a heterogeneous disease course. Insights into the genetics of the disease have identified certain high-risk cytogenetic features that are associated with adverse outcomes. While the advances in therapy have translated into dramatic improvements in the outcome of patients with MM, those with high-risk genetic features continue to perform poorly. This has resulted in a need for clinical trials focusing on the high-risk subgroup of MM as they search for additional biomarkers and therapeutic targets continue. In this review, we discuss the currently existing data on prognostic and predictive biomarkers in MM and speculate the role of treatment stratification based on the genetic features of the disease.
Collapse
Affiliation(s)
- Saurabh Zanwar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shaji Kumar
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
25
|
Su CT, Ye JC. Emerging therapies for relapsed/refractory multiple myeloma: CAR-T and beyond. J Hematol Oncol 2021; 14:115. [PMID: 34301270 PMCID: PMC8299593 DOI: 10.1186/s13045-021-01109-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023] Open
Abstract
The pace of innovation of multiple myeloma therapy in recent years is remarkable with the advent of monoclonal antibodies and the approval of novel agents with new mechanisms of action. Emerging therapies are on the horizon for clinical approval with significant implications in extending patient survival and advancing closer to the goal of a cure, especially in areas of immunotherapy such as chimeric antigen receptor T cells, bispecific T cell engager antibodies, antibody drug conjugates, newer generations of monoclonal antibodies, and small molecule inhibitor and modulators. This review provides an update of current myeloma therapeutics in active preclinical and early clinical development and discusses the mechanism of action of several classes of novel therapeutics.
Collapse
Affiliation(s)
- Christopher T Su
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - J Christine Ye
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
26
|
del(17p) without TP53 mutation confers a poor prognosis in intensively treated newly diagnosed patients with multiple myeloma. Blood 2021; 137:1192-1195. [PMID: 33080624 DOI: 10.1182/blood.2020008346] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Despite tremendous improvements in the outcome of patients with multiple myeloma in the past decade, high-risk patients have not benefited from the approval of novel drugs. The most important prognostic factor is the loss of parts of the short arm of chromosome 17, known as deletion 17p (del(17p)). A recent publication (on a small number of patients) suggested that these patients are at very high-risk only if del(17p) is associated with TP53 mutations, the so-called "double-hit" population. To validate this finding, we designed a much larger study on 121 patients presenting del(17p) in > 55% of their plasma cells, and homogeneously treated by an intensive approach. For these 121 patients, we performed deep next generation sequencing targeted on TP53. The outcome was then compared with a large control population (2505 patients lacking del(17p)). Our results confirmed that the "double hit" situation is the worst (median survival = 36 months), but that del(17p) alone also confers a poor outcome compared with the control cohort (median survival = 52.8 months vs 152.2 months, respectively). In conclusion, our study clearly confirms the extremely poor outcome of patients displaying "double hit," but also that del(17p) alone is still a very high-risk feature, confirming its value as a prognostic indicator for poor outcome.
Collapse
|
27
|
Zhu P, Qian T, Si C, Liu Y, Cui L, Huang W, Fu L, Deng C, Zeng T. High expression of CPNE5 and CPNE9 predicts positive prognosis in multiple myeloma. Cancer Biomark 2021; 31:77-85. [PMID: 33780365 DOI: 10.3233/cbm-203108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND CPNEs are significant biomarkers which can affect the progression and prognosis of various tumor diseases. However, the prognosis role of CPNEs in multiple myeloma (MM) is still unclear. OBJECTIVES To investigate the prognosis role of CPNEs in MM. METHODS Seven hundred and thirty-five samples from two independent data sets were involved to analyze the clinical and molecular characteristics, and prognosis role of the expression of CPNE1-9 in MM. RESULTS MM patients with higher expressions of CPNE5 and CPNE9 had longer event-free survival (EFS) and overall survival (OS) compared with CPNE5low and CPNE9low expression groups (EFS: P= 0.0054, 0.0065; OS: P= 0.015, 0.016, respectively). Multivariate regression analysis showed that CPNE5 was an independent favorable predictor for EFS and OS (EFS: P= 0.005; OS: P= 0.006), and CPNE9 was an independent positive indicator for EFS (P= 0.002). Moreover, the survival probability and the cumulative event of EFS and OS in CPNE5highCPNE9high group were significantly longer than other groups. CONCLUSIONS High expressions of CPNE5 and CPNE9 might be used as positive indicators for MM, and their combination was a better predictor for the survival of MM patients.
Collapse
Affiliation(s)
- Pei Zhu
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tingting Qian
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chaozeng Si
- Information Center, China-Japan Friendship Hospital, Beijing, China.,Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, Henan, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Longzhen Cui
- Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, Henan, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Wenhui Huang
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, Huaihe Hospital of Henan University, Kaifeng, Henan, China.,Department of Hematology, Huaihe Hospital of Henan University, Kaifeng, Henan, China
| | - Cong Deng
- Department of Clinical Laboratory, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tiansheng Zeng
- Department of Hematology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Translational Medicine Center, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumor Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
28
|
Risk factors in multiple myeloma: is it time for a revision? Blood 2021; 137:16-19. [PMID: 33024991 DOI: 10.1182/blood.2019004309] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022] Open
Abstract
Although therapeutic strategies have been adapted to age and comorbidities for a long time, almost all multiple myeloma (MM) patients currently receive similar treatment, whatever their disease risk category. However, high-risk MM patients still constitute an unmet medical need and should benefit from the most efficient drug combinations. Herein, we review and discuss how to optimally define risk and why a revision of the current definition is urgently needed.
Collapse
|
29
|
Lv M, Liu Q. JMJD2C triggers the growth of multiple myeloma cells via activation of β‑catenin. Oncol Rep 2021; 45:1162-1170. [PMID: 33469678 PMCID: PMC7860002 DOI: 10.3892/or.2021.7934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 12/02/2020] [Indexed: 01/10/2023] Open
Abstract
Emerging evidence has indicated that histone modification and its related regulators are involved in the progression of multiple myeloma (MM) cells. In the present study, the expression of Jumonji C domain‑containing 2 (JMJD2) was examined in both MM tissues and healthy controls. The roles of JMJD2C in the progression of MM were further investigated. The results revealed that the expression of JMJD2C, but not that of JMJD2A or JMJD2B, was increased in MM tissues compared with the healthy controls. The overexpression of JMJD2C significantly increased the in vitro growth of MM cells. The inhibitor of the β‑catenin signaling pathway significantly attenuated the JMJD2C‑induced growth of MM cells. Mechanistical analyses indicated that JMJD2C increased the transcription of β‑catenin in MM cells, which may be due to the fact that JMJD2C can directly bind with the promoter of β‑catenin. Furthermore, JMJD2C activated β‑catenin in MM cells via a GSK3β‑dependent manner, which was evidenced by the results demonstrating that the overexpression of GSK3β attenuated the JMJD2C‑induced decrease in the phosphorylation of β‑catenin. On the whole, the findings of the present study demonstrated that JMJD2C promotes the malignancy of MM via the activation of the β‑catenin pathway. These results suggested that JMJD2C may be a potential target for MM treatment.
Collapse
Affiliation(s)
- Ming Lv
- Department of Emergency Medicine, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277101, P.R. China
| | - Qicai Liu
- Department of Joint Surgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277101, P.R. China
| |
Collapse
|
30
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
31
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [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: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
32
|
Biallelic loss of BCMA as a resistance mechanism to CAR T cell therapy in a patient with multiple myeloma. Nat Commun 2021; 12:868. [PMID: 33558511 PMCID: PMC7870932 DOI: 10.1038/s41467-021-21177-5] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023] Open
Abstract
BCMA targeting chimeric antigen receptor (CAR) T cell therapy has shown deep and durable responses in multiple myeloma. However, relapse following therapy is frequently observed, and mechanisms of resistance remain ill-defined. Here, we perform single cell genomic characterization of longitudinal samples from a patient who relapsed after initial CAR T cell treatment with lack of response to retreatment. We report selection, following initial CAR T cell infusion, of a clone with biallelic loss of BCMA acquired by deletion of one allele and a mutation that creates an early stop codon on the second allele. This loss leads to lack of CAR T cell proliferation following the second infusion and is reflected by lack of soluble BCMA in patient serum. Our analysis suggests the need for careful detection of BCMA gene alterations in multiple myeloma cells from relapse following CAR T cell therapy. Relapse following BCMA targeted CAR T-cell therapy is frequently observed in patients with multiple myeloma (MM). Here, by single cell transcriptome profiling on serially collected bone marrow samples, the authors report biallelic loss of BCMA as the mechanism of resistance underlying both relapse and lack of response to a second CAR T infusion in a patient with MM.
Collapse
|
33
|
Lee N, Kim SM, Lee Y, Jeong D, Yun J, Ryu S, Yoon SS, Ahn YO, Hwang SM, Lee DS. Prognostic value of integrated cytogenetic, somatic variation, and copy number variation analyses in Korean patients with newly diagnosed multiple myeloma. PLoS One 2021; 16:e0246322. [PMID: 33544757 PMCID: PMC7864461 DOI: 10.1371/journal.pone.0246322] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/15/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND To investigate the prognostic value of gene variants and copy number variations (CNVs) in patients with newly diagnosed multiple myeloma (NDMM), an integrative genomic analysis was performed. METHODS Sixty-seven patients with NDMM exhibiting more than 60% plasma cells in the bone marrow aspirate were enrolled in the study. Whole-exome sequencing was conducted on bone marrow nucleated cells. Mutation and CNV analyses were performed using the CNVkit and Nexus Copy Number software. In addition, karyotype and fluorescent in situ hybridization were utilized for the integrated analysis. RESULTS Eighty-three driver gene mutations were detected in 63 patients with NDMM. The median number of mutations per patient was 2.0 (95% confidence interval [CI] = 2.0-3.0, range = 0-8). MAML2 and BHLHE41 mutations were associated with decreased survival. CNVs were detected in 56 patients (72.7%; 56/67). The median number of CNVs per patient was 6.0 (95% CI = 5.7-7.0; range = 0-16). Among the CNVs, 1q gain, 6p gain, 6q loss, 8p loss, and 13q loss were associated with decreased survival. Additionally, 1q gain and 6p gain were independent adverse prognostic factors. Increased numbers of CNVs and driver gene mutations were associated with poor clinical outcomes. Cluster analysis revealed that patients with the highest number of driver mutations along with 1q gain, 6p gain, and 13q loss exhibited the poorest prognosis. CONCLUSIONS In addition to the known prognostic factors, the integrated analysis of genetic variations and CNVs could contribute to prognostic stratification of patients with NDMM.
Collapse
Affiliation(s)
- Nuri Lee
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Sung-Min Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Youngeun Lee
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dajeong Jeong
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jiwon Yun
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sohee Ryu
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Clinical Research Institute, Seoul National University Hospital, Cancer Research Institute, Seoul National University, College of Medicine, Seoul, Korea
| | - Yong-Oon Ahn
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Mee Hwang
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Soon Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
34
|
A longitudinal analysis of chromosomal abnormalities in disease progression from MGUS/SMM to newly diagnosed and relapsed multiple myeloma. Ann Hematol 2021; 100:437-443. [PMID: 33392702 DOI: 10.1007/s00277-020-04384-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/16/2020] [Indexed: 12/30/2022]
Abstract
We analyzed variations in terms of chromosomal abnormalities (CA) by fluorescence in situ hybridization (FISH) analysis on purified bone marrow plasma cells throughout the progression from monoclonal gammopathy of undetermined significance/smoldering multiple myeloma (MGUS/SMM) to newly diagnosed MM/plasma cell leukemia (NDMM/PCL) at diagnosis and from diagnostic samples to progressive disease. High risk was defined by the presence of at least del(17p), t(4;14), and/or t(14;16). 1p/1q detection (in the standard FISH panel from 2012 onward) was not available for all patients. We analyzed 139 MM/PCL diagnostic samples from 144 patients, with a median follow-up of 71 months: high-risk CA at diagnosis (MGUS/SMM or NDMM) was present in 28% of samples, whereas 37-39% showed high-risk CA at relapse. In 115 patients with NDMM who evolved to relapsed/refractory MM, we identified 3 different populations: (1) 31/115 patients (27%) with gain of new CA (del13, del17p, t(4;14), t(14;16) or 1q CA when available); (2) 10/115 (9%) patients with loss of a previously identified CA; and (3) 74 patients with no changes. The CA gain group showed a median overall survival of 66 months vs. 84 months in the third group (HR 0.56, 95% CI 0.34-0.92, p = 0.023). Clonal evolution occurs as disease progresses after different chemotherapy lines. Patients who acquired high-risk CA had the poorest prognosis. Our findings highlight the importance of performing FISH analysis both at diagnosis and at relapse.
Collapse
|
35
|
Characterization of complete lncRNAs transcriptome reveals the functional and clinical impact of lncRNAs in multiple myeloma. Leukemia 2021; 35:1438-1450. [PMID: 33597729 PMCID: PMC8102198 DOI: 10.1038/s41375-021-01147-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/03/2020] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is an incurable disease, whose clinical heterogeneity makes its management challenging, highlighting the need for biological features to guide improved therapies. Deregulation of specific long non-coding RNAs (lncRNAs) has been shown in MM, nevertheless, the complete lncRNA transcriptome has not yet been elucidated. In this work, we identified 40,511 novel lncRNAs in MM samples. lncRNAs accounted for 82% of the MM transcriptome and were more heterogeneously expressed than coding genes. A total of 10,351 overexpressed and 9,535 downregulated lncRNAs were identified in MM patients when compared with normal bone-marrow plasma cells. Transcriptional dynamics study of lncRNAs in the context of normal B-cell maturation revealed 989 lncRNAs with exclusive expression in MM, among which 89 showed de novo epigenomic activation. Knockdown studies on one of these lncRNAs, SMILO (specific myeloma intergenic long non-coding RNA), resulted in reduced proliferation and induction of apoptosis of MM cells, and activation of the interferon pathway. We also showed that the expression of lncRNAs, together with clinical and genetic risk alterations, stratified MM patients into several progression-free survival and overall survival groups. In summary, our global analysis of the lncRNAs transcriptome reveals the presence of specific lncRNAs associated with the biological and clinical behavior of the disease.
Collapse
|
36
|
Locher M, Steurer M, Jukic E, Keller MA, Fresser F, Ruepp C, Wöll E, Verdorfer I, Gastl G, Willenbacher W, Weger R, Nachbaur D, Wolf D, Gunsilius E, Zschocke J, Steiner N. The prognostic value of additional copies of 1q21 in multiple myeloma depends on the primary genetic event. Am J Hematol 2020; 95:1562-1571. [PMID: 32936982 PMCID: PMC7756645 DOI: 10.1002/ajh.25994] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/19/2022]
Abstract
Hyperdiploidy (HRD) and specific immunoglobulin heavy locus (IGH) translocations are primary chromosomal abnormalities (CA) in multiple myeloma (MM). In this retrospective study of 794 MM patients we aimed to investigate clinical features and common CA including gain(1q) in separate subgroups defined by primary CA. In the entire group, we confirmed that gain(1q) was associated with short time to next treatment and adverse overall survival (OS). The impact was worse for four or more copies of 1q21 as compared to three copies. However, in a subgroup of patients with clonal gain(11q) and without known primary IGH translocations (CG11q), already three copies of 1q21 were associated with a poor outcome; in the absence of gain(1q), patients in this subgroup had a remarkably long median OS of more than nine years. These cases were associated with HRD, coexpression of CD56 and CD117, male gender, and IgG subtype. In non‐CG11q patients, four or more copies of 1q21 (but not three copies) had a significant adverse impact on outcome. Several associations with CA and clinical findings were observed for the defined subgroups. As an example, we found a predominance of early tetraploidy, plasma cell leukemia, and female gender in the t(14;16) subgroup. Our results underscore the importance of subgrouping in MM.
Collapse
Affiliation(s)
- Maurus Locher
- Institute of Human Genetics Medical University of Innsbruck Innsbruck Austria
| | - Michael Steurer
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
| | - Emina Jukic
- Institute of Human Genetics Medical University of Innsbruck Innsbruck Austria
| | - Markus A. Keller
- Institute of Human Genetics Medical University of Innsbruck Innsbruck Austria
| | - Friedrich Fresser
- Institute of Human Genetics Medical University of Innsbruck Innsbruck Austria
| | - Carmen Ruepp
- Department of Internal Medicine St. Vinzenz Krankenhaus Betriebs GmbH Zams Austria
| | - Ewald Wöll
- Department of Internal Medicine St. Vinzenz Krankenhaus Betriebs GmbH Zams Austria
| | - Irmgard Verdorfer
- Institute of Human Genetics Medical University of Innsbruck Innsbruck Austria
| | - Günther Gastl
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
| | - Wolfgang Willenbacher
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
- ONCOTYROL ‐ Center for Personalized Cancer Medicine Innsbruck Austria
| | - Roman Weger
- ONCOTYROL ‐ Center for Personalized Cancer Medicine Innsbruck Austria
| | - David Nachbaur
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
| | - Dominik Wolf
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
- Medical Clinic 3, Oncology, Hematology, Immuno‐Oncology and Rheumatology University Hospital Bonn Bonn Germany
| | - Eberhard Gunsilius
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
| | - Johannes Zschocke
- Institute of Human Genetics Medical University of Innsbruck Innsbruck Austria
| | - Normann Steiner
- Department of Internal Medicine V Medical University of Innsbruck Innsbruck Austria
| |
Collapse
|
37
|
Samur MK, Aktas Samur A, Fulciniti M, Szalat R, Han T, Shammas M, Richardson P, Magrangeas F, Minvielle S, Corre J, Moreau P, Thakurta A, Anderson KC, Parmigiani G, Avet-Loiseau H, Munshi NC. Genome-Wide Somatic Alterations in Multiple Myeloma Reveal a Superior Outcome Group. J Clin Oncol 2020; 38:3107-3118. [PMID: 32687451 PMCID: PMC7499613 DOI: 10.1200/jco.20.00461] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Multiple myeloma (MM) is accompanied by heterogeneous somatic alterations. The overall goal of this study was to describe the genomic landscape of myeloma using deep whole-genome sequencing (WGS) and develop a model that identifies patients with long survival. METHODS We analyzed deep WGS data from 183 newly diagnosed patients with MM treated with lenalidomide, bortezomib, and dexamethasone (RVD) alone or RVD + autologous stem cell transplant (ASCT) in the IFM/DFCI 2009 study (ClinicalTrials.gov identifier: NCT01191060). We integrated genomic markers with clinical data. RESULTS We report significant variability in mutational load and processes within MM subgroups. The timeline of observed activation of mutational processes provides the basis for 2 distinct models of acquisition of mutational changes detected at the time of diagnosis of myeloma. Virtually all MM subgroups have activated DNA repair-associated signature as a prominent late mutational process, whereas APOBEC signature targeting C>G is activated in the intermediate phase of disease progression in high-risk MM. Importantly, we identify a genomically defined MM subgroup (17% of newly diagnosed patients) with low DNA damage (low genomic scar score with chromosome 9 gain) and a superior outcome (100% overall survival at 69 months), which was validated in a large independent cohort. This subgroup allowed us to distinguish patients with low- and high-risk hyperdiploid MM and identify patients with prolongation of progression-free survival. Genomic characteristics of this subgroup included lower mutational load with significant contribution from age-related mutations as well as frequent NRAS mutation. Surprisingly, their overall survival was independent of International Staging System and minimal residual disease status. CONCLUSION This is a comprehensive study identifying genomic markers of a good-risk group with prolonged survival. Identification of this patient subgroup will affect future therapeutic algorithms and research planning.
Collapse
Affiliation(s)
- Mehmet Kemal Samur
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Anil Aktas Samur
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Raphael Szalat
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Tessa Han
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA
| | - Masood Shammas
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Paul Richardson
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Florence Magrangeas
- Inserm UMR892, CNRS 6299, Université de Nantes, and Centre Hospitalier Universitaire de Nantes, Unité Mixte de Genomique du Cancer, Nantes, France
| | - Stephane Minvielle
- Inserm UMR892, CNRS 6299, Université de Nantes, and Centre Hospitalier Universitaire de Nantes, Unité Mixte de Genomique du Cancer, Nantes, France
| | - Jill Corre
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Philippe Moreau
- Inserm UMR892, CNRS 6299, Université de Nantes, and Centre Hospitalier Universitaire de Nantes, Unité Mixte de Genomique du Cancer, Nantes, France
| | | | - Kenneth C. Anderson
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Giovanni Parmigiani
- Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Hervé Avet-Loiseau
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Nikhil C. Munshi
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
38
|
Ordoñez R, Kulis M, Russiñol N, Chapaprieta V, Carrasco-Leon A, García-Torre B, Charalampopoulou S, Clot G, Beekman R, Meydan C, Duran-Ferrer M, Verdaguer-Dot N, Vilarrasa-Blasi R, Soler-Vila P, Garate L, Miranda E, San José-Enériz E, Rodriguez-Madoz JR, Ezponda T, Martínez-Turrilas R, Vilas-Zornoza A, Lara-Astiaso D, Dupéré-Richer D, Martens JHA, El-Omri H, Taha RY, Calasanz MJ, Paiva B, San Miguel J, Flicek P, Gut I, Melnick A, Mitsiades CS, Licht JD, Campo E, Stunnenberg HG, Agirre X, Prosper F, Martin-Subero JI. Chromatin activation as a unifying principle underlying pathogenic mechanisms in multiple myeloma. Genome Res 2020; 30:1217-1227. [PMID: 32820006 PMCID: PMC7545147 DOI: 10.1101/gr.265520.120] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm associated with a broad variety of genetic lesions. In spite of this genetic heterogeneity, MMs share a characteristic malignant phenotype whose underlying molecular basis remains poorly characterized. In the present study, we examined plasma cells from MM using a multi-epigenomics approach and demonstrated that, when compared to normal B cells, malignant plasma cells showed an extensive activation of regulatory elements, in part affecting coregulated adjacent genes. Among target genes up-regulated by this process, we found members of the NOTCH, NF-kB, MTOR signaling, and TP53 signaling pathways. Other activated genes included sets involved in osteoblast differentiation and response to oxidative stress, all of which have been shown to be associated with the MM phenotype and clinical behavior. We functionally characterized MM-specific active distant enhancers controlling the expression of thioredoxin (TXN), a major regulator of cellular redox status and, in addition, identified PRDM5 as a novel essential gene for MM. Collectively, our data indicate that aberrant chromatin activation is a unifying feature underlying the malignant plasma cell phenotype.
Collapse
Affiliation(s)
- Raquel Ordoñez
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Marta Kulis
- Fundació Clínic per a la Recerca Biomèdica, 08036 Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Nuria Russiñol
- Fundació Clínic per a la Recerca Biomèdica, 08036 Barcelona, Spain
| | - Vicente Chapaprieta
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | - Beatriz García-Torre
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | | | - Guillem Clot
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Renée Beekman
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Cem Meydan
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, USA
| | - Martí Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Núria Verdaguer-Dot
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Roser Vilarrasa-Blasi
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Paula Soler-Vila
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Leire Garate
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Estíbaliz Miranda
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Edurne San José-Enériz
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | | | - Teresa Ezponda
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | | | - Amaia Vilas-Zornoza
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | - David Lara-Astiaso
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | - Daphné Dupéré-Richer
- Division of Hematology/Oncology, University of Florida Health Cancer Center, Gainesville, Florida 32610, USA
| | - Joost H A Martens
- Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, Netherlands
| | - Halima El-Omri
- Department of Hematology & BMT, Hamad Medical Corporation, NCCCR, Doha, Qatar
| | - Ruba Y Taha
- Department of Hematology & BMT, Hamad Medical Corporation, NCCCR, Doha, Qatar
| | - Maria J Calasanz
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Bruno Paiva
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jesus San Miguel
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton CB10 1SD, United Kingdom
| | - Ivo Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Ari Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York 10021, USA
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Jonathan D Licht
- Division of Hematology/Oncology, University of Florida Health Cancer Center, Gainesville, Florida 32610, USA
| | - Elias Campo
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Fundació Clínic per a la Recerca Biomèdica, 08036 Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Departamento de Fundamentos Clínicos, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | - Xabier Agirre
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Felipe Prosper
- Centro de Investigación Médica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Jose I Martin-Subero
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, 28029 Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Departamento de Fundamentos Clínicos, Universitat de Barcelona, 08036 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
39
|
Multiple Myeloma as a Bone Disease? The Tissue Disruption-Induced Cell Stochasticity (TiDiS) Theory. Cancers (Basel) 2020; 12:cancers12082158. [PMID: 32759688 PMCID: PMC7463431 DOI: 10.3390/cancers12082158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/21/2022] Open
Abstract
The standard model of multiple myeloma (MM) relies on genetic instability in the normal counterparts of MM cells. MM-induced lytic bone lesions are considered as end organ damages. However, bone is a tissue of significance in MM and bone changes could be at the origin/facilitate the emergence of MM. We propose the tissue disruption-induced cell stochasticity (TiDiS) theory for MM oncogenesis that integrates disruption of the microenvironment, differentiation, and genetic alterations. It starts with the observation that the bone marrow endosteal niche controls differentiation. As decrease in cellular stochasticity occurs thanks to cellular interactions in differentiating cells, the initiating role of bone disruption would be in the increase of cellular stochasticity. Thus, in the context of polyclonal activation of B cells, memory B cells and plasmablasts would compete for localizing in endosteal niches with the risk that some cells cannot fully differentiate if they cannot reside in the niche because of a disrupted microenvironment. Therefore, they would remain in an unstable state with residual proliferation, with the risk that subclones may transform into malignant cells. Finally, diagnostic and therapeutic perspectives are provided.
Collapse
|
40
|
Kanasugi J, Hanamura I, Ota A, Karnan S, Lam VQ, Mizuno S, Wahiduzzaman M, Rahman ML, Hyodo T, Konishi H, Tsuzuki S, Hosokawa Y, Takami A. Biallelic loss of FAM46C triggers tumor growth with concomitant activation of Akt signaling in multiple myeloma cells. Cancer Sci 2020; 111:1663-1675. [PMID: 32176823 PMCID: PMC7226186 DOI: 10.1111/cas.14386] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 02/21/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
Loss of heterozygosity or mutation of the family with sequence similarity 46, member C (FAM46C) gene on chromosome band 1p12 is associated with shorter overall survival of patients with multiple myeloma (MM). In this study, using human MM cell lines (KMS‐11, OCI‐My5, and ANBL‐6), we generated FAM46C−/− cell clones and examined the effect of disruption of FAM46C on cell survival and cellular signaling. Cell proliferation assays showed increased clonogenicity of FAM46C−/− KMS‐11 cells compared to WT cells. Xenograft experiments showed significantly shorter overall survival of mice harboring the FAM46C−/− cell‐derived tumors than mice with the FAM46CWT cell‐derived tumors. Notably, levels of phosphorylated Akt and its substrates increased both in vitro and in vivo in the FAM46C−/− cells compared to WT cells. In addition, caspase activities decreased in the FAM46C−/− cells. Results of gene set enrichment analysis showed that loss of FAM46C significantly activated serum‐responsive genes while inactivating phosphatase and tensin homolog (PTEN)‐related genes. Mechanistically, loss of FAM46C decreased the PTEN activity, number of apoptotic cells, and caspase activities. PF‐04691502, a selective PI3K inhibitor, suppressed the augmented phosphorylation of Akt and its substrate FoxO3a. Treatment with afuresertib (a specific Akt inhibitor) in combination with bortezomib additively decreased FAM46C−/− MM cell survival. Collectively, this study is the first to report that loss of FAM46C triggers the concomitant activation of the PI3K‐Akt signaling pathway, which might be a therapeutic target for MM with abnormalities in the FAM46C gene.
Collapse
Affiliation(s)
- Jo Kanasugi
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University, Nagakute, Japan
| | | | - Vu Quang Lam
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| | - Shohei Mizuno
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| | - Md Wahiduzzaman
- Department of Biochemistry, Aichi Medical University, Nagakute, Japan
| | - Md Lutfur Rahman
- Department of Biochemistry, Aichi Medical University, Nagakute, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University, Nagakute, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University, Nagakute, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University, Nagakute, Japan
| | | | - Akiyoshi Takami
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
41
|
Ferreira B, Caetano J, Barahona F, Lopes R, Carneiro E, Costa-Silva B, João C. Liquid biopsies for multiple myeloma in a time of precision medicine. J Mol Med (Berl) 2020; 98:513-525. [PMID: 32246161 PMCID: PMC7198642 DOI: 10.1007/s00109-020-01897-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a challenging, progressive, and highly heterogeneous hematological malignancy. MM is characterized by multifocal proliferation of neoplastic plasma cells in the bone marrow (BM) and sometimes in extramedullary organs. Despite the availability of novel drugs and the longer median overall survival, some patients survive more than 10 years while others die rapidly. This heterogeneity is mainly driven by biological characteristics of MM cells, including genetic abnormalities. Disease progressions are mainly due to the inability of drugs to overcome refractory disease and inevitable drug-resistant relapse. In clinical practice, a bone marrow biopsy, mostly performed in one site, is still used to access the genetics of MM. However, BM biopsy use is limited by its invasive nature and by often not accurately reflecting the mutational profile of MM. Recent insights into the genetic landscape of MM provide a valuable opportunity to implement precision medicine approaches aiming to enable better patient profiling and selection of targeted therapies. In this review, we explore the use of the emerging field of liquid biopsies in myeloma patients considering current unmet medical needs, such as assessing the dynamic mutational landscape of myeloma, early predictors of treatment response, and a less invasive response monitoring.
Collapse
Affiliation(s)
- Bruna Ferreira
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana Caetano
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Filipa Barahona
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Raquel Lopes
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Emilie Carneiro
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cristina João
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Nova Medical School, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
42
|
Long X, Xu Q, Lou Y, Li C, Gu J, Cai H, Wang D, Xu J, Li T, Zhou X, Xiao M, Wang Y, Mao X, Zhou J, Chen L. The utility of non-invasive liquid biopsy for mutational analysis and minimal residual disease assessment in extramedullary multiple myeloma. Br J Haematol 2020; 189:e45-e48. [PMID: 32191818 DOI: 10.1111/bjh.16440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Xiaolu Long
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Qian Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Yaoyao Lou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Jia Gu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Haodong Cai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Jinhuan Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Tongjuan Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Ying Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R.China
| |
Collapse
|
43
|
Abstract
Although therapeutic strategies have been adapted to age and comorbidities of myeloma patients for a long time, all patients currently experiment the same treatment whatever their genomic risk. However, high-risk patients should benefit right now from the most efficient drugs combinations. Herein, we review and discuss how to optimally define risk to adapt treatment and why a modern multiparametric definition of genomic risk is urgently needed. Minimal residual disease status will probably also take a growing place in patient's management, including in treatment adaptation. We also discuss how next-generation sequencing will definitively represent an essential tool to manage risk-based therapeutic strategies. Finally, despite an explosive knowledge of myeloma molecular landscape, targeted therapy perspectives remain poor, with only few exceptions.
Collapse
|
44
|
CNV Radar: an improved method for somatic copy number alteration characterization in oncology. BMC Bioinformatics 2020; 21:98. [PMID: 32143562 PMCID: PMC7060549 DOI: 10.1186/s12859-020-3397-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Cancer associated copy number variation (CNV) events provide important information for identifying patient subgroups and suggesting treatment strategies. Technical and logistical issues, however, make it challenging to accurately detect abnormal copy number events in a cost-effective manner in clinical studies. Results Here we present CNV Radar, a software tool that utilizes next-generation sequencing read depth information and variant allele frequency patterns, to infer the true copy number status of genes and genomic regions from whole exome sequencing data. Evaluation of CNV Radar in a public multiple myeloma dataset demonstrated that CNV Radar was able to detect a variety of CNVs associated with risk of progression, and we observed > 70% concordance with fluorescence in situ hybridization (FISH) results. Compared to other CNV callers, CNV Radar showed high sensitivity and specificity. Similar results were observed when comparing CNV Radar calls to single nucleotide polymorphism array results from acute myeloid leukemia and prostate cancer datasets available on TCGA. More importantly, CNV Radar demonstrated its utility in the clinical trial setting: in POLLUX and CASTOR, two phase 3 studies in patients with relapsed or refractory multiple myeloma, we observed a high concordance rate with FISH for del17p, a risk defining CNV event (88% in POLLUX and 90% in CASTOR), therefore allowing for efficacy assessments in clinically relevant disease subgroups. Our case studies also showed that CNV Radar is capable of detecting abnormalities such as copy-neutral loss of heterozygosity that elude other approaches. Conclusions We demonstrated that CNV Radar is more sensitive than other CNV detection methods, accurately detects clinically important cytogenetic events, and allows for further interrogation of novel disease biology. Overall, CNV Radar exhibited high concordance with standard methods such as FISH, and its success in the POLLUX and CASTOR clinical trials demonstrated its potential utility for informing clinical and therapeutic decisions.
Collapse
|
45
|
Wong KY, Chim CS. Venetoclax, bortezomib and S63845, an MCL1 inhibitor, in multiple myeloma. J Pharm Pharmacol 2020; 72:728-737. [DOI: 10.1111/jphp.13240] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 01/26/2020] [Indexed: 12/24/2022]
Abstract
Abstract
Objectives
Venetoclax, an orally available BCL2-selective inhibitor, has demonstrated promising single-agent anti-tumour activity in myeloma especially patients with t(11;14). Herein, whether venetoclax sensitivity could be enhanced or restored in combination with bortezomib or S63845, a novel MCL1-selective inhibitor, was examined in human myeloma cell lines (HMCLs), including bortezomib-resistant HMCLs.
Methods
By MTS assay, half-maximal inhibitory concentration (IC50) and hence sensitivity/resistance to venetoclax, bortezomib and S63845 were determined.
Key findings
Venetoclax (IC50 ≥100 nm), bortezomib (IC50 ≥50 nm) and S63845 (IC50 ≥100 nm) resistance was observed in nine (75%), three (25%) and six (50%) HMCLs, respectively. Moreover, venetoclax sensitivity was independent of bortezomib (R2 = 0.1107) or S63845 (R2 = 0.0213) sensitivity. Venetoclax sensitivity correlated with high mRNA ratio of BCL2/MCL1 (P = 0.0091), BCL2/BCL2L1 (P = 0.0182) and low MCL1 expression (P = 0.0091). In HMCLs sensitive to both venetoclax and bortezomib/S63845, venetoclax combined with S63845 showed stronger synergistic effect than combined with bortezomib. Moreover, in venetoclax-resistant HMCLs, S63845, but not bortezomib, significantly restored venetoclax sensitivity. Conversely, bortezomib combined with S63845 did not result in augmented bortezomib sensitivity or abolishment of bortezomib resistance.
Conclusions
Regardless of t(11;14), combination of venetoclax with S63845 is a promising strategy in enhancing venetoclax sensitivity or overcoming venetoclax resistance in myeloma therapy, hence warrant future clinical studies.
Collapse
Affiliation(s)
- Kwan Yeung Wong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
46
|
Hultcrantz M, Yellapantula V, Rustad EH. Genomic profiling of multiple myeloma: New insights and modern technologies. Best Pract Res Clin Haematol 2020; 33:101153. [PMID: 32139018 DOI: 10.1016/j.beha.2020.101153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023]
Abstract
Advances in technologies for genomic profiling, primarily with next generation sequencing, have lead to a better understanding of the complex genomic landscape in multiple myeloma. Integrated analysis of whole genome, exome and transcriptome sequencing has lead to new insights on disease drivers including translocations, copy number alterations, somatic mutations, and altered gene expression. Disease progression in multiple myeloma is largely driven by structural variations including the traditional immunoglobulin heavy chain (IGH) translocations and hyperdiploidy which are early events in myelomagenesis as well as more complex events spanning over multiple chromosomes and involving amplifications and deletions. In this review, we will discuss recent insights on the genomic landscape of multiple myeloma and their implications for disease progression and personalized treatment. We will review how sequencing assays compare to current clinical methods and give an overview of modern technologies for interrogating genomic aberrations.
Collapse
Affiliation(s)
- Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| | - Venkata Yellapantula
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Even H Rustad
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
47
|
Adamia S, Abiatari I, Amin SB, Fulciniti M, Minvielle S, Li C, Moreau P, Avet-Loiseau H, Munshi NC, Anderson KC. The effects of MicroRNA deregulation on pre-RNA processing network in multiple myeloma. Leukemia 2020; 34:167-179. [PMID: 31182781 PMCID: PMC6901818 DOI: 10.1038/s41375-019-0498-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/22/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022]
Abstract
Over the last few years, a detailed map of genetic and epigenetic lesions that underlie multiple myeloma (MM) has been created. Regulation of microRNA (miR)-dependent gene expression and mRNA splicing play significant roles in MM pathogenesis; however, to date an interplay between these processes is not yet delineated. Here we investigated miR-mediated regulation of splicing networks at the transcriptome level. Our studies show that a significant number (78%) of miRs which are either up- or down-regulated in patient CD138+ MM cells, but not in healthy donors (HD) CD138+ plasma cells (PC), target genes involved in early stages of pre-mRNA splicing. We also identified deregulated miRs that target core splicing factors (SF) and modifiers (SM, enhancers/silencers) which cause altered splicing in MM. Our studies suggest that Let-7f, in combination other miRs which are frequently and significantly deregulated in patients with overt MM, targets genes that regulate intron excision. Importantly, deregulated expression of certain miRs in MM promote increased intron retention, a novel characteristic of the MM genome, by inducing deregulated expression of the genes that regulate the splicing network. Our studies, therefore, provide the rationale for therapeutically targeting deregulated miRs to reverse aberrant splicing and improve patient outcome in MM.
Collapse
Affiliation(s)
- Sophia Adamia
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ivane Abiatari
- Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| | - Samir B Amin
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mariateresa Fulciniti
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Cheng Li
- Peking University, School of Life Sciences, Beijing, China
| | - Philippe Moreau
- Laboratory of Hematology, University Hospital, Nantes, France
| | | | - Nikhil C Munshi
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
48
|
Comprehensive detection of recurring genomic abnormalities: a targeted sequencing approach for multiple myeloma. Blood Cancer J 2019; 9:101. [PMID: 31827071 PMCID: PMC6906304 DOI: 10.1038/s41408-019-0264-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/31/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023] Open
Abstract
Recent genomic research efforts in multiple myeloma have revealed clinically relevant molecular subgroups beyond conventional cytogenetic classifications. Implementing these advances in clinical trial design and in routine patient care requires a new generation of molecular diagnostic tools. Here, we present a custom capture next-generation sequencing (NGS) panel designed to identify rearrangements involving the IGH locus, arm level, and focal copy number aberrations, as well as frequently mutated genes in multiple myeloma in a single assay. We sequenced 154 patients with plasma cell disorders and performed a head-to-head comparison with the results from conventional clinical assays, i.e., fluorescent in situ hybridization (FISH) and single-nucleotide polymorphism (SNP) microarray. Our custom capture NGS panel had high sensitivity (>99%) and specificity (>99%) for detection of IGH translocations and relevant chromosomal gains and losses in multiple myeloma. In addition, the assay was able to capture novel genomic markers associated with poor outcome such as bi-allelic events involving TP53. In summary, we show that a multiple myeloma designed custom capture NGS panel can detect IGH translocations and CNAs with very high concordance in relation to FISH and SNP microarrays and importantly captures the most relevant and recurrent somatic mutations in multiple myeloma rendering this approach highly suitable for clinical application in the modern era.
Collapse
|
49
|
Veenstra C, Bruce D, Timbs A, Hamblin A. Application of Genomics to Clinical Practice in Haematological Malignancy. CURRENT GENETIC MEDICINE REPORTS 2019. [DOI: 10.1007/s40142-019-00179-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
The usual abundance of fresh cells and high-quality DNA derived from bone marrow aspirate and peripheral blood mean haematological malignancies are at the forefront of the application of genomics to malignancy. This review evaluates where genomics is routinely used in clinical care and where opportunities for further application exist.
Recent Findings
The 2016 revision of the WHO classification of tumours of haematopoietic and lymphoid tissues increased the number of disease entities defined by, or whose diagnosis was strongly supported by, a specific genetic change. Increasingly combinations of mutations rather than individual lesions are being used to genomically classify heterogeneous disorders to inform prognosis and direct treatment. Furthermore, the role of different genetic aberrations as markers of measurable residual disease is being evaluated in clinical trials to allow intensification/de-intensification of treatment as appropriate and early detection of relapse.
Summary
Implementation of broader sequencing technologies such as whole exome/genome sequencing coupled with continuing developments in genomic technology to improve turn-around-times are likely to further reinforce the centrality of genomics in the management of haematological malignancies.
Collapse
|
50
|
Cypris O, Božić T, Wagner W. Chicken or Egg: Is Clonal Hematopoiesis Primarily Caused by Genetic or Epigenetic Aberrations? Front Genet 2019; 10:785. [PMID: 31552094 PMCID: PMC6746886 DOI: 10.3389/fgene.2019.00785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/24/2019] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic malignancies, including multiple myeloma, are associated with characteristic mutations and genetic instabilities that drive malignant transformation. On the other hand, tumor formation is also associated with drastic epigenetic aberrations, which can impact the genetic sequence. Therefore, the question arises if malignant transformation is primarily caused by genetic or epigenetic events. The tight connection of these processes becomes obvious by the fact that in several malignancies, as well as in age-related clonal hematopoiesis, mutations are particularly observed in epigenetic writers such as DNMT3A and TET2. On the other hand, specific epigenetic aberrations, so-called “epimutations,” can mimic genomic mutations. In contrast to the genetic sequence, which remains relatively stable throughout life, the epigenome notoriously undergoes drastic changes in normal hematopoietic development and aging. It is conceivable that such epigenetic reorganization, e.g., in 3D chromatin conformation, paves the way for secondary chromosomal instabilities, which then result in tumor-specific genomic changes that further trigger disease progression. This scenario might explain the occurrence of tumor-specific mutations particularly in the elderly. Taken together, the causality dilemma is difficult to solve because genetic and epigenetic aberrations are interlinked during disease development. A better understanding of how the chromatin structure or 3D nuclear organization can evoke specific mutations might provide new perspectives for prevention, early diagnostics, and targeted therapy.
Collapse
Affiliation(s)
- Olivia Cypris
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Engineering - Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Tanja Božić
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Engineering - Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany.,Institute for Biomedical Engineering - Cell Biology, University Hospital of RWTH Aachen, Aachen, Germany
| |
Collapse
|