351
|
Barrio S, DáVia M, Bruins L, Stühmer T, Steinbrunn T, Bittrich M, Einsele H, Stewart AK, Braggio E, Kortüm KM. Protocol for M 3P: A Comprehensive and Clinical Oriented Targeted Sequencing Panel for Routine Molecular Analysis in Multiple Myeloma. Methods Mol Biol 2018; 1792:117-128. [PMID: 29797255 DOI: 10.1007/978-1-4939-7865-6_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Over the past 10 years next generation sequencing (NGS) approaches deciphered a large number of genomes from a wide variety of tumor types. However, despite most relevant findings, this technology has not yet been implemented into standard diagnostic workflows. Broad access to NGS technology is still limited, sequencing/analysis times exceed clinically relevant timeframes and despite huge cuts, costs remain significant. We proposed a custom-tailored gene panel, which focuses on a selected number of relevant genes and developed a clinically oriented NGS targeted sequencing approach for the molecular characterization of Multiple Myeloma (MM) tumors, allowing the description of the tumor genetic heterogeneity and its changes under selective pressure of antitumor therapy, in a more cost effective and faster turnaround timeframe.
Collapse
Affiliation(s)
- Santiago Barrio
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Matteo DáVia
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Laura Bruins
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Thorsten Stühmer
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Torsten Steinbrunn
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Max Bittrich
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Alexander Keith Stewart
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Esteban Braggio
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - Klaus Martin Kortüm
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
352
|
Zhu S, Jin J, Gokhale S, Lu AM, Shan H, Feng J, Xie P. Genetic Alterations of TRAF Proteins in Human Cancers. Front Immunol 2018. [PMID: 30294322 DOI: 10.3389/fimmu.2018.02111/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
The tumor necrosis factor receptor (TNF-R)-associated factor (TRAF) family of cytoplasmic adaptor proteins regulate the signal transduction pathways of a variety of receptors, including the TNF-R superfamily, Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs), and cytokine receptors. TRAF-dependent signaling pathways participate in a diverse array of important cellular processes, including the survival, proliferation, differentiation, and activation of different cell types. Many of these TRAF-dependent signaling pathways have been implicated in cancer pathogenesis. Here we analyze the current evidence of genetic alterations of TRAF molecules available from The Cancer Genome Atlas (TCGA) and the Catalog of Somatic Mutations in Cancer (COSMIC) as well as the published literature, including copy number variations and mutation landscape of TRAFs in various human cancers. Such analyses reveal that both gain- and loss-of-function genetic alterations of different TRAF proteins are commonly present in a number of human cancers. These include pancreatic cancer, meningioma, breast cancer, prostate cancer, lung cancer, liver cancer, head and neck cancer, stomach cancer, colon cancer, bladder cancer, uterine cancer, melanoma, sarcoma, and B cell malignancies, among others. Furthermore, we summarize the key in vivo and in vitro evidence that demonstrates the causal roles of genetic alterations of TRAF proteins in tumorigenesis within different cell types and organs. Taken together, the information presented in this review provides a rationale for the development of therapeutic strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in different human cancers by precision medicine.
Collapse
Affiliation(s)
- Sining Zhu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Juan Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Pharmacology, Anhui Medical University, Hefei, China
| | - Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, NJ, United States
| | - Angeli M Lu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Haiyan Shan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jianjun Feng
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Engineering Research Center of the Modern Technology for Eel Industry, Ministry of Education of the People's Republic of China, Fisheries College of Jimei University, Xiamen, China
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
- Member, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
353
|
The Mutational Landscape of Circulating Tumor Cells in Multiple Myeloma. Cell Rep 2017; 19:218-224. [PMID: 28380360 PMCID: PMC5439509 DOI: 10.1016/j.celrep.2017.03.025] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 12/29/2016] [Accepted: 03/06/2017] [Indexed: 01/22/2023] Open
Abstract
The development of sensitive and non-invasive “liquid biopsies” presents new opportunities for longitudinal monitoring of tumor dissemination and clonal evolution. The number of circulating tumor cells (CTCs) is prognostic in multiple myeloma (MM), but there is little information on their genetic features. Here, we have analyzed the genomic landscape of CTCs from 29 MM patients, including eight cases with matched/paired bone marrow (BM) tumor cells. Our results show that 100% of clonal mutations in patient BM were detected in CTCs and that 99% of clonal mutations in CTCs were present in BM MM. These include typical driver mutations in MM such as in KRAS, NRAS, or BRAF. These data suggest that BM and CTC samples have similar clonal structures, as discordances between the two were restricted to subclonal mutations. Accordingly, our results pave the way for potentially less invasive mutation screening of MM patients through characterization of CTCs.
Collapse
|
354
|
Bustoros M, Mouhieddine TH, Detappe A, Ghobrial IM. Established and Novel Prognostic Biomarkers in Multiple Myeloma. Am Soc Clin Oncol Educ Book 2017; 37:548-560. [PMID: 28561668 DOI: 10.1200/edbk_175175] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy characterized by notable interpatient heterogeneity. There have been important advances in therapy and overall survival, but some patients with high-risk features still have poor survival rates. Therefore, accurate identification of this subset of patients has been integral to improvement of patient outcome. During the last few years, cytogenetics, gene expression profiling, MRI and PET/CT, as well as serum free light chain assays have been used as accurate biomarkers to better characterize the diverse course and outcome of the disease. With the recent advances of massive parallel sequencing techniques, the development of new models that better stratify high-risk groups are beginning to be developed. The use of multiparameter flow cytometry and next-generation sequencing have paved the way for assessment of minimal residual disease and better prognostication of post-therapeutic outcomes. Circulating tumor cells and circulating tumor DNA are promising potential biomarkers that demonstrate the spatial and temporal heterogeneity of MM. Finally, more prognostic markers are being developed that are specific to immunotherapeutic agents. In this review, we discuss these traditional and novel biomarkers that have been developed for MM and also those that can predict disease progression from precursor stages. Together, these biomarkers will help improve our understanding of the intrapatient and interpatient variabilities and help develop precision medicine for patients with high-risk MM.
Collapse
Affiliation(s)
- Mark Bustoros
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Tarek H Mouhieddine
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Alexandre Detappe
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Irene M Ghobrial
- From the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
355
|
Lionetti M, Barbieri M, Manzoni M, Fabris S, Bandini C, Todoerti K, Nozza F, Rossi D, Musto P, Baldini L, Neri A. Molecular spectrum of TP53 mutations in plasma cell dyscrasias by next generation sequencing: an Italian cohort study and overview of the literature. Oncotarget 2017; 7:21353-61. [PMID: 26870891 PMCID: PMC5008290 DOI: 10.18632/oncotarget.7241] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/19/2016] [Indexed: 12/30/2022] Open
Abstract
The prevalence of TP53 mutations greatly varies between tumor types; in multiple myeloma (MM) they were rarely detected at presentation, while increased frequency was reported with disease progression. Using next-generation sequencing, we analyzed TP53 exons 4-9 in a large representative cohort comprising patients with MM at diagnosis and more aggressive forms of plasma cell (PC) dyscrasia, identifying mutations in 4/129 (3%) MM, 6/24 (25%) primary PC leukemia, and 2/10 (20%) secondary PC leukemia cases. A similar increase in prevalence associated with disease aggressiveness (5%, 29.2% and 44%, respectively) was observed for TP53 deletion. Interestingly, in five patients mutations were not concomitant with TP53 deletion. Furthermore, longitudinal analysis revealed the acquisition of TP53 mutations in three of nineteen cases analyzed at relapse. Identified variants were mostly missense mutations concentrated in the DNA binding domain, only partly reflecting the pattern globally observed in human cancers. Our data confirm that TP53 mutations are rare in MM at presentation and rather represent a marker of progression, similarly to del(17p); however, their occurrence even in absence of deletions supports the importance of their assessment in patients with PC dyscrasia, in terms of both risk stratification and therapeutic implications.
Collapse
Affiliation(s)
- Marta Lionetti
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marzia Barbieri
- Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Martina Manzoni
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sonia Fabris
- Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Bandini
- Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Katia Todoerti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Filomena Nozza
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Davide Rossi
- Department of Translational Medicine, Division of Hematology, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Pellegrino Musto
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Luca Baldini
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Hematology Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
356
|
Ely S, Forsberg P, Ouansafi I, Rossi A, Modin A, Pearse R, Pekle K, Perry A, Coleman M, Jayabalan D, Di Liberto M, Chen-Kiang S, Niesvizky R, Mark TM. Cellular Proliferation by Multiplex Immunohistochemistry Identifies High-Risk Multiple Myeloma in Newly Diagnosed, Treatment-Naive Patients. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:825-833. [DOI: 10.1016/j.clml.2017.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 08/07/2017] [Accepted: 09/11/2017] [Indexed: 10/18/2022]
|
357
|
Stamato MA, Juli G, Romeo E, Ronchetti D, Arbitrio M, Caracciolo D, Neri A, Tagliaferri P, Tassone P, Amodio N. Inhibition of EZH2 triggers the tumor suppressive miR-29b network in multiple myeloma. Oncotarget 2017; 8:106527-106537. [PMID: 29290968 PMCID: PMC5739753 DOI: 10.18632/oncotarget.22507] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/29/2017] [Indexed: 11/25/2022] Open
Abstract
Downregulation of tumor suppressor (TS) microRNAs (miRNAs) commonly occurs in human cancer, including multiple myeloma (MM). We previously demonstrated that miR-29b is a relevant TS miRNA, whose expression in MM cells is inhibited by HDAC4-dependent deacetylation. Here, we provide novel insights into epigenetic mechanisms suppressing miR-29b in MM. In MM patient-derived plasma cells, we found inverse correlation between miR-29b and EZH2 mRNA expression. Both siRNAs and pharmacologic inhibitors of EZH2 led to miR-29b upregulation, and this effect was ascribed to reduced H3K27-trimethylation (H3K27me3) of miR-29a/b-1 promoter regions. Induction of miR-29b upon EZH2 inhibition occurred together with downregulation of major miR-29b pro-survival targets, such as SP1, MCL-1 and CDK6. Knock-down of the EZH2-interacting long non-coding RNA MALAT1 also reduced H3K27me3 of miR-29a/b-1 promoter, along with induction of miR-29b and downregulation of miR-29b targets. Importantly, inhibition of miR-29b by antagomiRs dramatically reduced in vitro anti-MM activity of small molecule EZH2-inhibitors, indicating that functional miR-29b is crucial for the activity of these compounds. Altogether, these results disclose novel epigenetic alterations contributing to the suppression of miR-29b molecular network, which can be instrumental for the development of rationally designed miRNA-based anti-MM therapeutics.
Collapse
Affiliation(s)
- Maria Angelica Stamato
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Enrica Romeo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Domenica Ronchetti
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, US
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy
| |
Collapse
|
358
|
Insights on Genomic and Molecular Alterations in Multiple Myeloma and Their Incorporation towards Risk-Adapted Treatment Strategy: Concise Clinical Review. Int J Genomics 2017; 2017:6934183. [PMID: 29250532 PMCID: PMC5698810 DOI: 10.1155/2017/6934183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/10/2017] [Indexed: 11/23/2022] Open
Abstract
Although recent advances in novel treatment approaches and therapeutics have shifted the treatment landscape of multiple myeloma, it remains an incurable plasma cell malignancy. Growing knowledge of the genome and expressed genomic information characterizing the biologic behavior of multiple myeloma continues to accumulate. However, translation and incorporation of vast molecular understanding of complex tumor biology to deliver personalized and precision treatment to cure multiple myeloma have not been successful to date. Our review focuses on current evidence and understanding of myeloma biology with characterization in the context of genomic and molecular alterations. We also discuss future clinical application of the genomic and molecular knowledge, and more translational research is needed to benefit our myeloma patients.
Collapse
|
359
|
Shah V, Boyd KD, Houlston RS, Kaiser MF. Constitutional mutation in CDKN2A is associated with long term survivorship in multiple myeloma: a case report. BMC Cancer 2017; 17:718. [PMID: 29110637 PMCID: PMC5674776 DOI: 10.1186/s12885-017-3715-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/30/2017] [Indexed: 11/11/2022] Open
Abstract
Background Multiple Myeloma is a cancer of plasma cells associated with significantly reduced survival. Long term survivorship from myeloma is very rare and despite advances in its treatment the disease is generally considered incurable. We report a patient diagnosed with myeloma carrying a germline mutation of a tumour suppressor gene who has effectively been cured. Case presentation A 36-year-old woman was diagnosed with IgG lambda myeloma in 1985. She was treated with melphalan chemotherapy followed by high-dose melphalan and autologous stem cell rescue and since remained in complete remission despite not having received any additional therapy. After eliciting a prior history of multiple primary melanomas and breast cancer, she was tested for and shown to be a carrier for a germline mutation in CDKN2A. Conclusions This is the second case report of germline mutation of CDKN2A being associated with myeloma. CDKN2A is a stabiliser of p53. Long term survivorship after high dose DNA damaging chemotherapy with melphalan in this patient is compatible with an increased chemo-sensitivity due to impairment of the DNA repair pathway.
Collapse
Affiliation(s)
- Vallari Shah
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
| | - Kevin D Boyd
- Department of Haemato-Oncology, Royal Marsden Hospital, London, UK
| | - Richard S Houlston
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.,Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Martin F Kaiser
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
360
|
Identification of gene pairs through penalized regression subject to constraints. BMC Bioinformatics 2017; 18:466. [PMID: 29100492 PMCID: PMC5670721 DOI: 10.1186/s12859-017-1872-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023] Open
Abstract
Background This article concerns the identification of gene pairs or combinations of gene pairs associated with biological phenotype or clinical outcome, allowing for building predictive models that are not only robust to normalization but also easily validated and measured by qPCR techniques. However, given a small number of biological samples yet a large number of genes, this problem suffers from the difficulty of high computational complexity and imposes challenges to the accuracy of identification statistically. Results In this paper, we propose a parsimonious model representation and develop efficient algorithms for identification. Particularly, we derive an equivalent model subject to a sum-to-zero constraint in penalized linear regression, where the correspondence between nonzero coefficients in these models is established. Most importantly, it reduces the model complexity of the traditional approach from the quadratic order to the linear order in the number of candidate genes, while overcoming the difficulty of model nonidentifiablity. Computationally, we develop an algorithm using the alternating direction method of multipliers (ADMM) to deal with the constraint. Numerically, we demonstrate that the proposed method outperforms the traditional method in terms of the statistical accuracy. Moreover, we demonstrate that our ADMM algorithm is more computationally efficient than a coordinate descent algorithm with a local search. Finally, we illustrate the proposed method on a prostate cancer dataset to identify gene pairs that are associated with pre-operative prostate-specific antigen. Conclusion Our findings demonstrate the feasibility and utility of using gene pairs as biomarkers.
Collapse
|
361
|
Clonal Evolution in Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 16 Suppl:S130-4. [PMID: 27521309 DOI: 10.1016/j.clml.2016.02.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/09/2016] [Indexed: 01/03/2023]
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy encountered among patients in the United States. The last decade has seen incremental improvements in the survival of patients with MM. These advances are, to a large extent, attributable to the addition of proteasome inhibitors and immunomodulatory drugs to the armamentarium of treatment options. The adoption of these drug classes was the result of an empiric research paradigm. However, with the application of next generation sequencing technologies, we are now starting to unravel the genomic landscape of MM. It is hoped that this will allow us to better disentangle the biology of the disease and allow for identification of new therapeutic targets. In this article, we review what we have learned to date about the mutational profile, clonal architecture, and evolution of the disease, and discuss the potential clinical implications of these findings.
Collapse
|
362
|
Manojlovic Z, Christofferson A, Liang WS, Aldrich J, Washington M, Wong S, Rohrer D, Jewell S, Kittles RA, Derome M, Auclair D, Craig DW, Keats J, Carpten JD. Comprehensive molecular profiling of 718 Multiple Myelomas reveals significant differences in mutation frequencies between African and European descent cases. PLoS Genet 2017; 13:e1007087. [PMID: 29166413 PMCID: PMC5699827 DOI: 10.1371/journal.pgen.1007087] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/23/2017] [Indexed: 12/30/2022] Open
Abstract
Multiple Myeloma (MM) is a plasma cell malignancy with significantly greater incidence and mortality rates among African Americans (AA) compared to Caucasians (CA). The overall goal of this study is to elucidate differences in molecular alterations in MM as a function of self-reported race and genetic ancestry. Our study utilized somatic whole exome, RNA-sequencing, and correlated clinical data from 718 MM patients from the Multiple Myeloma Research Foundation CoMMpass study Interim Analysis 9. Somatic mutational analyses based upon self-reported race corrected for ancestry revealed significant differences in mutation frequency between groups. Of interest, BCL7A, BRWD3, and AUTS2 demonstrate significantly higher mutation frequencies among AA cases. These genes are all involved in translocations in B-cell malignancies. Moreover, we detected a significant difference in mutation frequency of TP53 and IRF4 with frequencies higher among CA cases. Our study provides rationale for interrogating diverse tumor cohorts to best understand tumor genomics across populations.
Collapse
Affiliation(s)
- Zarko Manojlovic
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | | | - Winnie S. Liang
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Jessica Aldrich
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Megan Washington
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Shukmei Wong
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Daniel Rohrer
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | - Scott Jewell
- Van Andel Research Institute, Grand Rapids, MI, United States of America
| | - Rick A. Kittles
- Department of Surgery, Division of Population Genetics, University of Arizona, Tuscon, AZ, United States of America
| | - Mary Derome
- Multiple Myeloma Research Foundation, Norwalk, CT, United States of America
| | - Daniel Auclair
- Multiple Myeloma Research Foundation, Norwalk, CT, United States of America
| | - David Wesley Craig
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Jonathan Keats
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - John D. Carpten
- Department of Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| |
Collapse
|
363
|
Abstract
PURPOSE OF REVIEW Next generation sequencing and large-scale analysis of patient specimens has created a more complete picture of multiple myeloma (MM) revealing that epigenetic deregulation is a prominent factor in MM pathogenesis. RECENT FINDINGS Over half of MM patients have mutations in genes encoding epigenetic modifier enzymes. The DNA methylation profile of MM is related to the stage of the disease and certain classes of mutations in epigenetic modifiers are more prevalent upon disease relapse, suggesting a role in disease progression. Many small molecules targeting regulators of epigenetic machinery have been developed and clinical trials are underway for some of these in MM. SUMMARY Recent findings suggest that epigenetic targeting drugs could be an important strategy to cure MM. Combining these agents along with other strategies to affect the MM cell such as immunomodulatory drugs and proteasome inhibitors may enhance efficacy of combination regimens in MM.
Collapse
|
364
|
Abstract
PURPOSE OF REVIEW New risk stratification systems and treatment strategies have been introduced in recent years. We aim to provide an overview of these recent changes and summarise these data in a concise article that would be useful for clinicians. RECENT FINDINGS Apart from clinical stage, disease genetics are now recognised as important prognostic risk factors, and various new cytogenetic changes with negative prognostic impact have been identified. New technologies such as minimal residual disease detection are also playing an important role in prognostic assessment. Recent introduction of combination therapy with proteasome inhibitors and immunomodulatory drugs is showing promising results in high-risk patients and may partially abrogate the negative impact associated with some of the adverse risk factors. Recent advance has improved our understanding of high-risk multiple myeloma, and new therapeutic agents are now coming through the pipeline for this patient group with once dismal outcome.
Collapse
Affiliation(s)
- Henry S H Chan
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON, M5G 2M9, Canada
| | - Christine I Chen
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON, M5G 2M9, Canada
| | - Donna E Reece
- Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON, M5G 2M9, Canada.
| |
Collapse
|
365
|
Mailankody S, Kazandjian D, Korde N, Roschewski M, Manasanch E, Bhutani M, Tageja N, Kwok M, Zhang Y, Zingone A, Lamy L, Costello R, Morrison C, Hultcrantz M, Christofferson A, Washington M, Boateng M, Steinberg SM, Stetler-Stevenson M, Figg WD, Papaemmanuil E, Wilson WH, Keats JJ, Landgren O. Baseline mutational patterns and sustained MRD negativity in patients with high-risk smoldering myeloma. Blood Adv 2017; 1:1911-1918. [PMID: 29296837 PMCID: PMC5728141 DOI: 10.1182/bloodadvances.2017005934] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 08/14/2017] [Indexed: 02/07/2023] Open
Abstract
Early results of a prospective phase 2 clinical trial of carfilzomib, lenalidomide, and dexamethasone followed by lenalidomide maintenance in high-risk smoldering myeloma showed promising results that were previously published. Here, we provide novel insights into the genetic landscape of high-risk smoldering myeloma and information on sustained minimal residual disease (MRD) negativity with an expanded cohort of patients. Eighteen patients with high-risk smoldering myeloma were enrolled between 29 May 2012, and 14 January 2014. We included patients with newly diagnosed multiple myeloma enrolled in a parallel trial who received the same therapy (reference group). The overall response rate was 100%. With median potential follow-up of 43.3 months, 10 (63%) remain in MRD negativity, and the estimated 4-year progression-free and overall survival rates are 71% and 100%, respectively. Importantly, we report differences in mutational patterns in patients with high-risk smoldering myeloma and newly diagnosed multiple myeloma, reflected in a lower frequency of mutations in significant myeloma genes (6.6% vs 45%) and NFKB pathway genes (6.6% vs 25%). Treatment with carfilzomib, lenalidomide, and dexamethasone followed by lenalidomide maintenance was associated with a 100% response rate and 63% MRD negativity with a safety profile consistent with previous reports for this regimen. This study had a small numbers of participants, but there seemed to be important differences in the genetic landscape of patients with high-risk smoldering myeloma and those with newly diagnosed multiple myeloma, suggestive of a more treatment-responsive biology in early disease.
Collapse
Affiliation(s)
- Sham Mailankody
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dickran Kazandjian
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Neha Korde
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mark Roschewski
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Elisabet Manasanch
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Manisha Bhutani
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC
| | - Nishant Tageja
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mary Kwok
- Hematology/Oncology Department, John P. Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, MD
| | - Yong Zhang
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Adriana Zingone
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laurence Lamy
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rene Costello
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Candis Morrison
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Division of Hematology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | - Megan Washington
- Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, AZ
| | - Martin Boateng
- Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, AZ
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; and
| | | | - William D Figg
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Elli Papaemmanuil
- Computational Oncology, Epidemiology and Biostatics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wyndham H Wilson
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jonathan J Keats
- Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, AZ
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
366
|
Mithraprabhu S, Spencer A. Circulating tumour DNA analysis in multiple myeloma. Oncotarget 2017; 8:90610-90611. [PMID: 29207579 PMCID: PMC5710860 DOI: 10.18632/oncotarget.21595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Sridurga Mithraprabhu
- Andrew Spencer: Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| | - Andrew Spencer
- Andrew Spencer: Australian Centre for Blood Diseases, Monash University, Malignant Haematology & Stem Cell Transplantation, Alfred Hospital and Department of Clinical Haematology, Monash University, Melbourne, Australia
| |
Collapse
|
367
|
Clinical utility of recently identified diagnostic, prognostic, and predictive molecular biomarkers in mature B-cell neoplasms. Mod Pathol 2017; 30:1338-1366. [PMID: 28664939 DOI: 10.1038/modpathol.2017.58] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/18/2022]
Abstract
Genomic profiling studies have provided new insights into the pathogenesis of mature B-cell neoplasms and have identified markers with prognostic impact. Recurrent mutations in tumor-suppressor genes (TP53, BIRC3, ATM), and common signaling pathways, such as the B-cell receptor (CD79A, CD79B, CARD11, TCF3, ID3), Toll-like receptor (MYD88), NOTCH (NOTCH1/2), nuclear factor-κB, and mitogen activated kinase signaling, have been identified in B-cell neoplasms. Chronic lymphocytic leukemia/small lymphocytic lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, Waldenström macroglobulinemia, hairy cell leukemia, and marginal zone lymphomas of splenic, nodal, and extranodal types represent examples of B-cell neoplasms in which novel molecular biomarkers have been discovered in recent years. In addition, ongoing retrospective correlative and prospective outcome studies have resulted in an enhanced understanding of the clinical utility of novel biomarkers. This progress is reflected in the 2016 update of the World Health Organization classification of lymphoid neoplasms, which lists as many as 41 mature B-cell neoplasms (including provisional categories). Consequently, molecular genetic studies are increasingly being applied for the clinical workup of many of these neoplasms. In this review, we focus on the diagnostic, prognostic, and/or therapeutic utility of molecular biomarkers in mature B-cell neoplasms.
Collapse
|
368
|
Miller A, Asmann Y, Cattaneo L, Braggio E, Keats J, Auclair D, Lonial S, Russell SJ, Stewart AK. High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma. Blood Cancer J 2017; 7:e612. [PMID: 28937974 PMCID: PMC5709757 DOI: 10.1038/bcj.2017.94] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 06/29/2017] [Indexed: 12/11/2022] Open
Abstract
Tumor-specific mutations can result in immunogenic neoantigens, both of which have been correlated with responsiveness to immune checkpoint inhibitors in highly mutagenic cancers. However, early results of single-agent checkpoint inhibitors in multiple myeloma (MM) have been underwhelming. Therefore, we sought to understand the relationship between mutation and neoantigen landscape of MM patients and responsiveness to therapies. Somatic mutation burden, neoantigen load, and response to therapy were determined using interim data from the MMRF CoMMpass study (NCT01454297) on 664 MM patients. In this population, the mean somatic and missense mutation loads were 405.84(s=608.55) and 63.90(s=95.88) mutations per patient, respectively. There was a positive linear relationship between mutation and neoantigen burdens (R2=0.862). The average predicted neoantigen load was 23.52(s=52.14) neoantigens with an average of 9.40(s=26.97) expressed neoantigens. Survival analysis revealed significantly shorter progression-free survival (PFS) in patients with greater than average somatic missense mutation load (N=163, 0.493 vs 0.726 2-year PFS, P=0.0023) and predicted expressed neoantigen load (N=214, 0.555 vs 0.729 2-year PFS, P=0.0028). This pattern is maintained when stratified by disease stage and cytogenetic abnormalities. Therefore, high mutation and neoantigen load are clinically relevant risk factors that negatively impact survival of MM patients under current standards of care.
Collapse
Affiliation(s)
- A Miller
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Y Asmann
- Division of Biomedical Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | - L Cattaneo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - E Braggio
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - J Keats
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - D Auclair
- Multiple Myeloma Research Foundation, Norwalk CT, USA
| | - S Lonial
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA
| | | | - S J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Hematology, Mayo Clinic Rochester, MN, USA
| | - A K Stewart
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
369
|
|
370
|
van Beers EH, van Vliet MH, Kuiper R, de Best L, Anderson KC, Chari A, Jagannath S, Jakubowiak A, Kumar SK, Levy JB, Auclair D, Lonial S, Reece D, Richardson P, Siegel DS, Stewart AK, Trudel S, Vij R, Zimmerman TM, Fonseca R. Prognostic Validation of SKY92 and Its Combination With ISS in an Independent Cohort of Patients With Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:555-562. [DOI: 10.1016/j.clml.2017.06.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/08/2017] [Indexed: 10/19/2022]
|
371
|
Abstract
The outcomes for the majority of patients with myeloma have improved over recent decades, driven by treatment advances. However, there is a subset of patients considered to have high-risk disease who have not benefited. Understanding how high-risk disease evolves from more therapeutically tractable stages is crucial if we are to improve outcomes. This can be accomplished by identifying the genetic mechanisms and mutations driving the transition of a normal plasma cell to one with the features of the following disease stages: monoclonal gammopathy of undetermined significance, smouldering myeloma, myeloma and plasma cell leukaemia. Although myeloma initiating events are clonal, subsequent driver lesions often occur in a subclone of cells, facilitating progression by Darwinian selection processes. Understanding the co-evolution of the clones within their microenvironment will be crucial for therapeutically manipulating the process. The end stage of progression is the generation of a state associated with treatment resistance, increased proliferation, evasion of apoptosis and an ability to grow independently of the bone marrow microenvironment. In this Review, we discuss these end-stage high-risk disease states and how new information is improving our understanding of their evolutionary trajectories, how they may be diagnosed and the biological behaviour that must be addressed if they are to be treated effectively.
Collapse
Affiliation(s)
- Charlotte Pawlyn
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | - Gareth J Morgan
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| |
Collapse
|
372
|
Logic programming reveals alteration of key transcription factors in multiple myeloma. Sci Rep 2017; 7:9257. [PMID: 28835615 PMCID: PMC5569101 DOI: 10.1038/s41598-017-09378-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/25/2017] [Indexed: 01/24/2023] Open
Abstract
Innovative approaches combining regulatory networks (RN) and genomic data are needed to extract biological information for a better understanding of diseases, such as cancer, by improving the identification of entities and thereby leading to potential new therapeutic avenues. In this study, we confronted an automatically generated RN with gene expression profiles (GEP) from a cohort of multiple myeloma (MM) patients and normal individuals using global reasoning on the RN causality to identify key-nodes. We modeled each patient by his or her GEP, the RN and the possible automatically detected repairs needed to establish a coherent flow of the information that explains the logic of the GEP. These repairs could represent cancer mutations leading to GEP variability. With this reasoning, unmeasured protein states can be inferred, and we can simulate the impact of a protein perturbation on the RN behavior to identify therapeutic targets. We showed that JUN/FOS and FOXM1 activities are altered in almost all MM patients and identified two survival markers for MM patients. Our results suggest that JUN/FOS-activation has a strong impact on the RN in view of the whole GEP, whereas FOXM1-activation could be an interesting way to perturb an MM subgroup identified by our method.
Collapse
|
373
|
Neutral tumor evolution in myeloma is associated with poor prognosis. Blood 2017; 130:1639-1643. [PMID: 28827410 DOI: 10.1182/blood-2016-11-750612] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 08/07/2017] [Indexed: 12/24/2022] Open
Abstract
Recent studies suggest that the evolutionary history of a cancer is important in forecasting clinical outlook. To gain insight into the clonal dynamics of multiple myeloma (MM) and its possible influence on patient outcomes, we analyzed whole exome sequencing tumor data for 333 patients from Myeloma XI, a UK phase 3 trial and 434 patients from the CoMMpass study, all of which had received immunomodulatory drug (IMiD) therapy. By analyzing mutant allele frequency distributions in tumors, we found that 17% to 20% of MM is under neutral evolutionary dynamics. These tumors are associated with poorer patient survival in nonintensively treated patients, which is consistent with the reduced therapeutic efficacy of microenvironment-modulating IMiDs. Our findings provide evidence that knowledge of the evolutionary history of MM has relevance for predicting patient outcomes and personalizing therapy.
Collapse
|
374
|
Spatial genomic heterogeneity in multiple myeloma revealed by multi-region sequencing. Nat Commun 2017; 8:268. [PMID: 28814763 PMCID: PMC5559527 DOI: 10.1038/s41467-017-00296-y] [Citation(s) in RCA: 283] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 06/20/2017] [Indexed: 12/27/2022] Open
Abstract
In multiple myeloma malignant plasma cells expand within the bone marrow. Since this site is well-perfused, a rapid dissemination of "fitter" clones may be anticipated. However, an imbalanced distribution of multiple myeloma is frequently observed in medical imaging. Here, we perform multi-region sequencing, including iliac crest and radiology-guided focal lesion specimens from 51 patients to gain insight into the spatial clonal architecture. We demonstrate spatial genomic heterogeneity in more than 75% of patients, including inactivation of CDKN2C and TP53, and mutations affecting mitogen-activated protein kinase genes. We show that the extent of spatial heterogeneity is positively associated with the size of biopsied focal lesions consistent with regional outgrowth of advanced clones. The results support a model for multiple myeloma progression with clonal sweeps in the early phase and regional evolution in advanced disease. We suggest that multi-region investigations are critical to understanding intra-patient heterogeneity and the evolutionary processes in multiple myeloma.In multiple myeloma, malignant cells expand within bone marrow. Here, the authors use multi-region sequencing in patient samples to analyse spatial clonal architecture and heterogeneity, providing novel insight into multiple myeloma progression and evolution.
Collapse
|
375
|
Chillón MC, Jiménez C, García-Sanz R, Alcoceba M, Prieto I, García-Alvarez M, Antón A, Maldonado R, Hernández-Ruano M, González M, Gutiérrez NC, Sarasquete ME. Quantitative PCR: an alternative approach to detect common copy number alterations in multiple myeloma. Ann Hematol 2017; 96:1699-1705. [PMID: 28770277 DOI: 10.1007/s00277-017-3083-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022]
Abstract
Chromosome 1q gains and 13q deletions are common cytogenetic aberrations in multiple myeloma (MM) that confer a poor prognosis. There are several techniques for the targeted study of these alterations, but interphase fluorescence in situ hybridization (FISH) is the current gold standard. The aim of the present study was to validate quantitative PCR (qPCR) as an alternative to FISH studies in CD138+-enriched plasma cells (PCs) from MM patients at diagnosis. We analyzed 1q gains and 13q deletions by qPCR in 57 and 60 MM patients, respectively. qPCR applicability was 84 and 88% for 1q and 13q, respectively. The qPCR and FISH methods had a sensitivity and specificity of 88 and 71% for 1q gains, and 79 and 100% for 13q deletions. A second qPCR assay for each region was carried out to confirm the previous results. Paired qPCR (two assays) and FISH results were available from 53 MM patients: 26 for 1q amplification and 27 for 13q deletion. qPCR assays gave concordant results (qPCR-consistent) in 20 of the 26 (77%) 1q gains and 25 of the 27 (93%) 13q deletions. Considering only the consistent data, the overall concordance among qPCR and FISH was 85 and 100% for 1q gains and 13q deletions, respectively. Our results show a substantial agreement between qPCR and the gold standard FISH technique, indicating the potential of qPCR as an alternative approach, particularly when the starting material is too scarce or cells are too damaged to obtain accurate results from FISH studies.
Collapse
Affiliation(s)
- M C Chillón
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,CIBERONC, Madrid, Spain
| | - C Jiménez
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - R García-Sanz
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,CIBERONC, Madrid, Spain.
| | - M Alcoceba
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,CIBERONC, Madrid, Spain
| | - I Prieto
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - M García-Alvarez
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - A Antón
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - R Maldonado
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - M Hernández-Ruano
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - M González
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,CIBERONC, Madrid, Spain
| | - N C Gutiérrez
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain
| | - M E Sarasquete
- Hospital Universitario de Salamanca-Instituto de Investigación Biomédica de Salamanca (IBSAL), Centro de Investigación del Cáncer-IBMCC-CSIC, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.,CIBERONC, Madrid, Spain
| |
Collapse
|
376
|
Shah GL, Landau H, Londono D, Devlin SM, Kosuri S, Lesokhin AM, Lendvai N, Hassoun H, Chung DJ, Koehne G, Jhanwar SC, Landgren O, Levine R, Giralt SA. Gain of chromosome 1q portends worse prognosis in multiple myeloma despite novel agent-based induction regimens and autologous transplantation. Leuk Lymphoma 2017; 58:1823-1831. [PMID: 28078910 PMCID: PMC5554510 DOI: 10.1080/10428194.2016.1260126] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
We aimed to identify whether the use of autologous hematopoietic cell transplantation (HCT) impacts outcomes for multiple myeloma patients with gains of chromosome 1q (+1q). We retrospectively identified 95 patients, 21% having +1q. For patients with +1q, the overall response rate to induction was 85%, with 40% having ≥ VGPR and 20% achieving a CR, similar to non +1q patients (p = .64). The median PFS from diagnosis with +1q was 2.1 years (95% CI: 1.2-not reached (NR)) vs 4.3 years (95% CI: 3.3 yrs-NR) without +1q (p = .003). Median OS from diagnosis was 4.4 years (95% CI: 2.9-NR) vs not reached, respectively (p = .005). On molecular analysis using the Foundation One Heme assay, the most common mutations seen in +1q patients included TP53 (38%) and KRAS (25%). Overall, gain of 1q portends worse PFS and OS which was not negated by auto HCT. Such patients will likely require additional therapy to improve their survival.
Collapse
Affiliation(s)
- Gunjan L. Shah
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Heather Landau
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Dory Londono
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean M. Devlin
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Satyajit Kosuri
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander M. Lesokhin
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Nikoletta Lendvai
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Hani Hassoun
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - David J. Chung
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Guenther Koehne
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Suresh C. Jhanwar
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Ross Levine
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| | - Sergio A. Giralt
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, NY Presbyterian Hospital – Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
377
|
Tahara K, Takizawa M, Yamane A, Osaki Y, Ishizaki T, Mitsui T, Yokohama A, Saitoh T, Tsukamoto N, Matsumoto M, Murakami H, Nojima Y, Handa H. Overexpression of B-cell lymphoma 6 alters gene expression profile in a myeloma cell line and is associated with decreased DNA damage response. Cancer Sci 2017; 108:1556-1564. [PMID: 28544233 PMCID: PMC5543477 DOI: 10.1111/cas.13283] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/22/2017] [Accepted: 05/10/2017] [Indexed: 01/02/2023] Open
Abstract
B-cell lymphoma 6 (BCL6) attenuates DNA damage response (DDR) through gene repression and facilitates tolerance to genomic instability during immunoglobulin affinity maturation in germinal center (GC) B cells. Although BCL6 expression is repressed through normal differentiation of GC B cells into plasma cells, a recent study showed the ectopic expression of BCL6 in primary multiple myeloma (MM) cells. However, the functional roles of BCL6 in MM cells are largely unknown. Here, we report that overexpression of BCL6 in a MM cell line, KMS12PE, induced transcriptional repression of ataxia telangiectasia mutated (ATM), a DDR signaling kinase, which was associated with a reduction in γH2AX formation after DNA damage. In contrast, transcription of known targets of BCL6 in GC B cells was not affected, suggesting a cell type-specific function of BCL6. To further investigate the effects of BCL6 overexpression on the MM cell line, we undertook mRNA sequence analysis and found an upregulation in the genomic mutator activation-induced cytidine deaminase (AID) with alteration in the gene expression profile, which is suggestive of de-differentiation from plasma cells. Moreover, interleukin-6 exposure to KMS12PE led to upregulation of BCL6 and AID, downregulation of ATM, and attenuation of DDR, which were consistent with the effects of BCL6 overexpression in this MM cell line. Taken together, these results indicated that overexpression of BCL6 alters gene expression profile and confers decreased DDR in MM cells. This phenotypic change could be reproduced by interleukin-6 stimulation, suggesting an important role of external stimuli in inducing genomic instability, which is a hallmark of MM cells.
Collapse
Affiliation(s)
- Kenichi Tahara
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Makiko Takizawa
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Arito Yamane
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Yohei Osaki
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Takuma Ishizaki
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Takeki Mitsui
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Akihiko Yokohama
- Division of Blood Transfusion ServiceGunma University HospitalGunmaJapan
| | - Takayuki Saitoh
- Department of Laboratory SciencesGunma University Graduate School of Health SciencesGunmaJapan
| | | | - Morio Matsumoto
- Department of HematologyNational Hospital Organization Nishigunma National HospitalGunmaJapan
| | - Hirokazu Murakami
- Department of Laboratory SciencesGunma University Graduate School of Health SciencesGunmaJapan
| | - Yoshihisa Nojima
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| | - Hiroshi Handa
- Department of Medicine and Clinical ScienceGunma University Graduate School of MedicineGunmaJapan
| |
Collapse
|
378
|
Abstract
Multiple myeloma is a malignancy of terminally differentiated plasma cells, and patients typically present with bone marrow infiltration of clonal plasma cells and monoclonal protein in the serum and/or urine. The diagnosis of multiple myeloma is made when clear end-organ damage attributable to the plasma cell proliferative disorder or when findings that suggest a high likelihood of their development are present. Distinguishing symptomatic multiple myeloma that requires treatment from the precursor stages of monoclonal gammopathy of undetermined significance and smouldering multiple myeloma is important, as observation is the standard for those conditions. Much progress has been made over the past decade in the understanding of disease biology and individualized treatment approaches. Several new classes of drugs, such as proteasome inhibitors and immunomodulatory drugs, have joined the traditional armamentarium (corticosteroids, alkylating agents and anthracyclines) and, along with high-dose therapy and autologous haemopoietic stem cell transplantation, have led to deeper and durable clinical responses. Indeed, an increasing proportion of patients are achieving lasting remissions, raising the possibility of cure for this disease. Success will probably depend on using combinations of effective agents and treating patients in the early stages of disease, such as patients with smouldering multiple myeloma.
Collapse
|
379
|
Abstract
OBJECTIVES This integrative review describes the genomic variants that have been found to be associated with poor prognosis in patients diagnosed with multiple myeloma (MM). Second, it identifies MM genetic and genomic changes using next-generation sequencing, specifically whole-genome sequencing or exome sequencing. DATA SOURCE A search for peer-reviewed articles through PubMed, EBSCOhost, and DePaul WorldCat Libraries Worldwide yielded 33 articles that were included in the final analysis. CONCLUSION The most commonly reported genetic changes were KRAS, NRAS, TP53, FAM46C, BRAF, DIS3, ATM, and CCND1. These genetic changes play a role in the pathogenesis of MM, prognostication, and therapeutic targets for novel therapies. IMPLICATIONS FOR NURSING PRACTICE MM genetics and genomics are expanding rapidly; oncology nurse clinicians must have basic competencies in genetics and genomics to help patients understand the complexities of genetic and genomic alterations and be able to refer patients to appropriate genomic professionals if needed.
Collapse
|
380
|
Kiss KP, Varga G, Mikala G, Balassa K, Bors A, Kovy P, Meggyesi N, Kozma A, Csacsovszki O, Remenyi P, Valyi-Nagy I, Tordai A, Masszi T, Andrikovics H. The adverse effect of FOPNL genomic variant is reversed by bortezomib-based treatment protocols in multiple myeloma. Leuk Lymphoma 2017; 59:710-716. [PMID: 28691553 DOI: 10.1080/10428194.2017.1346250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Fibroblast growth factor receptor 1 oncogene partner N-terminal like gene (FOPNL) rs72773978 polymorphism was identified as an adverse prognostic factor in multiple myeloma (MM). We aimed to investigate the associations of rs72773978 with clinical characteristics and treatment outcome in 373 Hungarian MM patients. In our cohort, FOPNL polymorphism showed differential prognostic effect that depended on the treatment applied. Among patients treated with non-proteasome inhibitor (PI)-based therapy, carriership of the minor allele was significantly associated with adverse overall survival (p=.022). In contrast, the adverse effect was overcome by the application of PI-containing treatment (p=.048). Multivariate analyses revealed the independent adverse effect of rs72773978 on survival in the non-PI-treated group (p=.045), but not in PI treatment (OS: p=.093). We confirmed the adverse prognostic effect of rs72773978 associated with non-PI-based treatment regimens. Our results point to the importance of genotypic prognostic information associated with complex clinical background MM.
Collapse
Affiliation(s)
- Katalin Piroska Kiss
- a Laboratory of Molecular Diagnostics , Hungarian National Blood Transfusion Service , Budapest , Hungary
| | - Gergely Varga
- b 3rd Department of Internal Medicine , Semmelweis University , Budapest , Hungary
| | - Gabor Mikala
- c Department of Haematology and Stem Cell Transplantation , St. Istvan and St. Laszlo Hospital , Budapest , Hungary
| | - Katalin Balassa
- a Laboratory of Molecular Diagnostics , Hungarian National Blood Transfusion Service , Budapest , Hungary
| | - Andras Bors
- a Laboratory of Molecular Diagnostics , Hungarian National Blood Transfusion Service , Budapest , Hungary
| | - Petra Kovy
- a Laboratory of Molecular Diagnostics , Hungarian National Blood Transfusion Service , Budapest , Hungary
| | - Nora Meggyesi
- a Laboratory of Molecular Diagnostics , Hungarian National Blood Transfusion Service , Budapest , Hungary
| | - Andras Kozma
- c Department of Haematology and Stem Cell Transplantation , St. Istvan and St. Laszlo Hospital , Budapest , Hungary
| | - Otto Csacsovszki
- c Department of Haematology and Stem Cell Transplantation , St. Istvan and St. Laszlo Hospital , Budapest , Hungary
| | - Peter Remenyi
- c Department of Haematology and Stem Cell Transplantation , St. Istvan and St. Laszlo Hospital , Budapest , Hungary
| | - Istvan Valyi-Nagy
- c Department of Haematology and Stem Cell Transplantation , St. Istvan and St. Laszlo Hospital , Budapest , Hungary
| | - Attila Tordai
- d Department of Pathophysiology , Semmelweis University , Budapest , Hungary
| | - Tamas Masszi
- b 3rd Department of Internal Medicine , Semmelweis University , Budapest , Hungary.,c Department of Haematology and Stem Cell Transplantation , St. Istvan and St. Laszlo Hospital , Budapest , Hungary
| | - Hajnalka Andrikovics
- a Laboratory of Molecular Diagnostics , Hungarian National Blood Transfusion Service , Budapest , Hungary
| |
Collapse
|
381
|
Tisato V, Voltan R, Gonelli A, Secchiero P, Zauli G. MDM2/X inhibitors under clinical evaluation: perspectives for the management of hematological malignancies and pediatric cancer. J Hematol Oncol 2017; 10:133. [PMID: 28673313 PMCID: PMC5496368 DOI: 10.1186/s13045-017-0500-5] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023] Open
Abstract
The two murine double minute (MDM) family members MDM2 and MDMX are at the center of an intense clinical assessment as molecular target for the management of cancer. Indeed, the two proteins act as regulators of P53, a well-known key controller of the cell cycle regulation and cell proliferation that, when altered, plays a direct role on cancer development and progression. Several evidence demonstrated that functional aberrations of P53 in tumors are in most cases the consequence of alterations on the MDM2 and MDMX regulatory proteins, in particular in patients with hematological malignancies where TP53 shows a relatively low frequency of mutation while MDM2 and MDMX are frequently found amplified/overexpressed. The pharmacological targeting of these two P53-regulators in order to restore or increase P53 expression and activity represents therefore a strategy for cancer therapy. From the discovery of the Nutlins in 2004, several compounds have been developed and reported with the ability of targeting the P53-MDM2/X axis by inhibiting MDM2 and/or MDMX. From natural compounds up to small molecules and stapled peptides, these MDM2/X pharmacological inhibitors have been extensively studied, revealing different biological features and different rate of efficacy when tested in in vitro and in vivo experimental tumor models. The data/evidence coming from the preclinical experimentation have allowed the identification of the most promising molecules and the setting of clinical studies for their evaluation as monotherapy or in therapeutic combination with conventional chemotherapy or with innovative therapeutic protocols in different tumor settings. Preliminary results have been recently published reporting data about safety, tolerability, potential side effects, and efficacy of such therapeutic approaches. In this light, the aim of this review is to give an updated overview about the state of the art of the clinical evaluation of MDM2/X inhibitor compounds with a special attention to hematological malignancies and to the potential for the management of pediatric cancers.
Collapse
Affiliation(s)
- Veronica Tisato
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy.
| | - Rebecca Voltan
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| |
Collapse
|
382
|
The therapeutic potential of cell cycle targeting in multiple myeloma. Oncotarget 2017; 8:90501-90520. [PMID: 29163849 PMCID: PMC5685770 DOI: 10.18632/oncotarget.18765] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/11/2017] [Indexed: 12/15/2022] Open
Abstract
Proper cell cycle progression through the interphase and mitosis is regulated by coordinated activation of important cell cycle proteins (including cyclin-dependent kinases and mitotic kinases) and several checkpoint pathways. Aberrant activity of these cell cycle proteins and checkpoint pathways results in deregulation of cell cycle progression, which is one of the key hallmarks of cancer. Consequently, intensive research on targeting these cell cycle regulatory proteins identified several candidate small molecule inhibitors that are able to induce cell cycle arrest and even apoptosis in cancer cells. Importantly, several of these cell cycle regulatory proteins have also been proposed as therapeutic targets in the plasma cell malignancy multiple myeloma (MM). Despite the enormous progress in the treatment of MM the past 5 years, MM still remains most often incurable due to the development of drug resistance. Deregulated expression of the cyclins D is observed in virtually all myeloma patients, emphasizing the potential therapeutic interest of cyclin-dependent kinase inhibitors in MM. Furthermore, other targets have also been identified in MM, such as microtubules, kinesin motor proteins, aurora kinases, polo-like kinases and the anaphase promoting complex/cyclosome. This review will provide an overview of the cell cycle proteins and checkpoint pathways deregulated in MM and discuss the therapeutic potential of targeting proteins or protein complexes involved in cell cycle control in MM.
Collapse
|
383
|
González-Calle V, Keane N, Braggio E, Fonseca R. Precision Medicine in Myeloma: Challenges in Defining an Actionable Approach. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2017; 17:621-630. [PMID: 28743429 DOI: 10.1016/j.clml.2017.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 06/08/2017] [Indexed: 12/18/2022]
Abstract
Recently, large sequencing studies have provided insights into the mutational landscape of multiple myeloma (MM), identifying actionable mutations and giving a precious opportunity for exploring new targeted therapies. The main goal of precision medicine, matching patients with the right drug, seems to be closer than ever. However, no targeted therapies in MM are approved yet. Several clinical trials testing targeted drugs and enrolling patients with MM are currently ongoing and will provide predictive biomarkers that might support clinical decision making. In this review, we evaluate the evidence supporting the implementation of precision medicine in MM and we discuss the challenges that should be dealt with in this imminent and promising new era.
Collapse
Affiliation(s)
| | - Niamh Keane
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ
| | - Esteban Braggio
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ
| | - Rafael Fonseca
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ.
| |
Collapse
|
384
|
Zhu YX, Shi CX, Bruins LA, Jedlowski P, Wang X, Kortüm KM, Luo M, Ahmann JM, Braggio E, Stewart AK. Loss of FAM46C Promotes Cell Survival in Myeloma. Cancer Res 2017; 77:4317-4327. [PMID: 28619709 DOI: 10.1158/0008-5472.can-16-3011] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/17/2017] [Accepted: 06/09/2017] [Indexed: 02/06/2023]
Abstract
FAM46C is one of the most recurrently mutated genes in multiple myeloma; however its role in disease pathogenesis has not been determined. Here we demonstrate that wild-type (WT) FAM46C overexpression induces substantial cytotoxicity in multiple myeloma cells. In contrast, FAM46C mutations found in multiple myeloma patients abrogate this cytotoxicity, indicating a survival advantage conferred by the FAM46C mutant phenotype. WT FAM46C overexpression downregulated IRF4, CEBPB, and MYC and upregulated immunoglobulin (Ig) light chain and HSPA5/BIP Furthermore, pathway analysis suggests that enforced FAM46C expression activated the unfolded protein response pathway and induced mitochondrial dysfunction. CRISPR-mediated depletion of endogenous FAM46C enhanced multiple myeloma cell growth, decreased Ig light chain and HSPA5/BIP expression, activated ERK and antiapoptotic signaling, and conferred relative resistance to dexamethasone and lenalidomide treatments. Genes altered in FAM46C-depleted cells were enriched for signaling pathways regulating estrogen, glucocorticoid, B-cell receptor signaling, and ATM signaling. Together these results implicate FAM46C in myeloma cell growth and survival and identify FAM46C mutation as a contributor to myeloma pathogenesis and disease progression via perturbation in plasma cell differentiation and endoplasmic reticulum homeostasis. Cancer Res; 77(16); 4317-27. ©2017 AACR.
Collapse
Affiliation(s)
- Yuan Xiao Zhu
- Division of Hematology, Mayo Clinic Scottsdale, Arizona
| | - Chang-Xin Shi
- Division of Hematology, Mayo Clinic Scottsdale, Arizona
| | | | | | - Xuewei Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic Rochester, Minnesota
| | | | - Moulun Luo
- Center for Metabolic and Vascular Biology, Arizona State University, Tempe, Arizona
| | | | | | - A Keith Stewart
- Division of Hematology, Mayo Clinic Scottsdale, Arizona. .,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
385
|
Diagnosis and classification of hematologic malignancies on the basis of genetics. Blood 2017; 130:410-423. [PMID: 28600336 DOI: 10.1182/blood-2017-02-734541] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Genomic analysis has greatly influenced the diagnosis and clinical management of patients affected by diverse forms of hematologic malignancies. Here, we review how genetic alterations define subclasses of patients with acute leukemias, myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPNs), non-Hodgkin lymphomas, and classical Hodgkin lymphoma. These include new subtypes of acute myeloid leukemia defined by mutations in RUNX1 or BCR-ABL1 translocations as well as a constellation of somatic structural DNA alterations in acute lymphoblastic leukemia. Among patients with MDS, detection of mutations in SF3B1 define a subgroup of patients with the ring sideroblast form of MDS and a favorable prognosis. For patients with MPNs, detection of the BCR-ABL1 fusion delineates chronic myeloid leukemia from classic BCR-ABL1- MPNs, which are largely defined by mutations in JAK2, CALR, or MPL In the B-cell lymphomas, detection of characteristic rearrangements involving MYC in Burkitt lymphoma, BCL2 in follicular lymphoma, and MYC/BCL2/BCL6 in high-grade B-cell lymphomas are essential for diagnosis. In T-cell lymphomas, anaplastic large-cell lymphoma is defined by mutually exclusive rearrangements of ALK, DUSP22/IRF4, and TP63 Genetic alterations affecting TP53 and the mutational status of the immunoglobulin heavy-chain variable region are important in clinical management of chronic lymphocytic leukemia. Additionally, detection of BRAFV600E mutations is helpful in the diagnosis of classical hairy cell leukemia and a number of histiocytic neoplasms. Numerous additional examples provided here demonstrate how clinical evaluation of genomic alterations have refined classification of myeloid neoplasms and major forms of lymphomas arising from B, T, or natural killer cells.
Collapse
|
386
|
Shah V, Sherborne AL, Walker BA, Johnson DC, Boyle EM, Ellis S, Begum DB, Proszek PZ, Jones JR, Pawlyn C, Savola S, Jenner MW, Drayson MT, Owen RG, Houlston RS, Cairns DA, Gregory WM, Cook G, Davies FE, Jackson GH, Morgan GJ, Kaiser MF. Prediction of outcome in newly diagnosed myeloma: a meta-analysis of the molecular profiles of 1905 trial patients. Leukemia 2017; 32:102-110. [PMID: 28584253 PMCID: PMC5590713 DOI: 10.1038/leu.2017.179] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/11/2017] [Accepted: 05/25/2017] [Indexed: 12/11/2022]
Abstract
Robust establishment of survival in multiple myeloma (MM) and its relationship to recurrent genetic aberrations is required as outcomes are variable despite apparent similar staging. We assayed copy number alterations (CNA) and translocations in 1036 patients from the NCRI Myeloma XI trial and linked these to overall survival (OS) and progression-free survival. Through a meta-anlysis of these data with data from MRC Myeloma IX trial, totalling 1905 newly diagnosed MM patients (NDMM), we confirm the association of t(4;14), t(14;16), t(14;20), del(17p) and gain(1q21) with poor prognosis with hazard ratios (HRs) for OS of 1.60 (P=4.77 × 10-7), 1.74 (P=0.0005), 1.90 (P=0.0089), 2.10 (P=8.86 × 10-14) and 1.68 (P=2.18 × 10-14), respectively. Patients with 'double-hit' defined by co-occurrence of at least two adverse lesions have an especially poor prognosis with HRs for OS of 2.67 (P=8.13 × 10-27) for all patients and 3.19 (P=1.23 × 10-18) for intensively treated patients. Using comprehensive CNA and translocation profiling in Myeloma XI we also demonstrate a strong association between t(4;14) and BIRC2/BIRC3 deletion (P=8.7 × 10-15), including homozygous deletion. Finally, we define distinct sub-groups of hyperdiploid MM, with either gain(1q21) and CCND2 overexpression (P<0.0001) or gain(11q25) and CCND1 overexpression (P<0.0001). Profiling multiple genetic lesions can identify MM patients likely to relapse early allowing stratification of treatment.
Collapse
Affiliation(s)
- V Shah
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - A L Sherborne
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - B A Walker
- MIRT, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - D C Johnson
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - E M Boyle
- Department of Haematology, Hopital Huriez, CHRU, Lille, France
| | - S Ellis
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - D B Begum
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - P Z Proszek
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - J R Jones
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - C Pawlyn
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - S Savola
- MRC Holland, Amsterdam, The Netherlands
| | - M W Jenner
- Department of Haematology, University Hospital Southampton, Southampton, UK
| | - M T Drayson
- Clinical Immunology, School of Immunity & Infection, University of Birmingham, Birmingham, UK
| | - R G Owen
- Department of Haematology, St James's University Hospital, Leeds, UK
| | - R S Houlston
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - D A Cairns
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | - W M Gregory
- Clinical Trials Research Unit, University of Leeds, Leeds, UK
| | - G Cook
- Section of Experimental Haematology, Leeds Institute of Cancer & Pathology, University of Leeds, Leeds, UK
| | - F E Davies
- MIRT, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - G H Jackson
- Department of Haematology, Newcastle University, Newcastle upon Tyne, UK
| | - G J Morgan
- MIRT, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - M F Kaiser
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
387
|
Lionetti M, Neri A. Utilizing next-generation sequencing in the management of multiple myeloma. Expert Rev Mol Diagn 2017; 17:653-663. [PMID: 28524737 DOI: 10.1080/14737159.2017.1332996] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Multiple myeloma (MM) is a bone marrow plasma cell malignancy characterized by wide clinical presentation and heterogeneous genetic background. Despite the recent advances in patient outcome, new markers are needed for improving risk prediction and choice of a more appropriate therapy. In this perspective, the genetic makeup of MM cells is being better characterized by means of next-generation sequencing (NGS) technologies. Areas covered: The authors discuss how the application of NGS has improved our knowledge of MM biology by discovering its mutational landscape, identifying the operating mutational processes, and revealing the clonal composition of tumors and the dynamics of its evolution; and how this can have important clinical implications in terms of prognostication, therapeutic choices, and response assessment. Finally, the authors provide a quick outlook of future applications of these technologies that could help in the management of the disease in the next years. Expert commentary: The clinical exploitation of NGS-based characterization of MM patients has as its ultimate goal the precision medicine. Considerable obstacles to its implementation in myeloma management exist; therefore, the concerted effort of all involved stakeholders is mandatory to ensure that it will become a reality in routine clinical practice in the next future.
Collapse
Affiliation(s)
- Marta Lionetti
- a Department of Oncology and Hemato-oncology , Università degli Studi di Milano , Milano , Italy.,b Hematology , Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Antonino Neri
- a Department of Oncology and Hemato-oncology , Università degli Studi di Milano , Milano , Italy.,b Hematology , Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico , Milano , Italy
| |
Collapse
|
388
|
Mikulasova A, Wardell CP, Murison A, Boyle EM, Jackson GH, Smetana J, Kufova Z, Pour L, Sandecka V, Almasi M, Vsianska P, Gregora E, Kuglik P, Hajek R, Davies FE, Morgan GJ, Walker BA. The spectrum of somatic mutations in monoclonal gammopathy of undetermined significance indicates a less complex genomic landscape than that in multiple myeloma. Haematologica 2017; 102:1617-1625. [PMID: 28550183 PMCID: PMC5685224 DOI: 10.3324/haematol.2017.163766] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/16/2017] [Indexed: 01/10/2023] Open
Abstract
Monoclonal gammopathy of undetermined significance is a pre-malignant precursor of multiple myeloma with a 1% risk of progression per year. Although targeted analyses have shown the presence of specific genetic abnormalities such as IGH translocations, RB1 deletion, 1q gain, hyperdiploidy or RAS gene mutations, little is known about the molecular mechanism of malignant transformation. We performed whole exome sequencing together with comparative genomic hybridization plus single nucleotide polymorphism array analysis in 33 flow-cytometry-separated abnormal plasma cell samples from patients with monoclonal gammopathy of undetermined significance to describe somatic gene mutations and chromosome changes at the genome-wide level. Non-synonymous mutations and copy-number alterations were present in 97.0% and in 60.6% of cases, respectively. Importantly, the number of somatic mutations was significantly lower in monoclonal gammopathy of undetermined significance than in myeloma (P<10-4) and we identified six genes that were significantly mutated in myeloma (KRAS, NRAS, DIS3, HIST1H1E, EGR1 and LTB) within the monoclonal gammopathy of undetermined significance dataset. We also found a positive correlation with increasing chromosome changes and somatic gene mutations. IGH translocations, comprising t(4;14), t(11;14), t(14;16) and t(14;20), were present in 27.3% of cases and in a similar frequency to myeloma, consistent with the primary lesion hypothesis. MYC translocations and TP53 deletions or mutations were not detected in samples from patients with monoclonal gammopathy of undetermined significance, indicating that they may be drivers of progression to myeloma. Data from this study show that monoclonal gammopathy of undetermined significance is genetically similar to myeloma, however overall genetic abnormalities are present at significantly lower levels in monoclonal gammopathy of undetermined significant than in myeloma.
Collapse
Affiliation(s)
- Aneta Mikulasova
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Medical Genetics, University Hospital Brno, Czech Republic.,Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Christopher P Wardell
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Alexander Murison
- Center for Myeloma Research, Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Eileen M Boyle
- Center for Myeloma Research, Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Graham H Jackson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Jan Smetana
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Medical Genetics, University Hospital Brno, Czech Republic
| | - Zuzana Kufova
- Faculty of Medicine, University of Ostrava, Czech Republic.,Department of Hematooncology, University Hospital Ostrava, Czech Republic
| | - Ludek Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Czech Republic
| | - Viera Sandecka
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Czech Republic
| | - Martina Almasi
- Department of Clinical Hematology, University Hospital Brno, Czech Republic
| | - Pavla Vsianska
- Department of Clinical Hematology, University Hospital Brno, Czech Republic
| | - Evzen Gregora
- Department of Internal Medicine and Hematology, University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Petr Kuglik
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,Department of Medical Genetics, University Hospital Brno, Czech Republic
| | - Roman Hajek
- Faculty of Medicine, University of Ostrava, Czech Republic.,Department of Hematooncology, University Hospital Ostrava, Czech Republic
| | - Faith E Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian A Walker
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
389
|
Bolli N, Barcella M, Salvi E, D'Avila F, Vendramin A, De Philippis C, Munshi NC, Avet-Loiseau H, Campbell PJ, Mussetti A, Carniti C, Maura F, Barlassina C, Corradini P, Montefusco V. Next-generation sequencing of a family with a high penetrance of monoclonal gammopathies for the identification of candidate risk alleles. Cancer 2017; 123:3701-3708. [DOI: 10.1002/cncr.30777] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Niccolo Bolli
- Department of Oncology and Hemato-Oncology; University of Milan; Milan Italy
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
- The Cancer Genome Project; Wellcome Trust Sanger Institute; Cambridge United Kingdom
| | - Matteo Barcella
- Genomic and Bioinformatics Unit; Department of Health Sciences, University of Milan; Milan Italy
| | - Erika Salvi
- Genomic and Bioinformatics Unit; Department of Health Sciences, University of Milan; Milan Italy
| | - Francesca D'Avila
- Genomic and Bioinformatics Unit; Department of Health Sciences, University of Milan; Milan Italy
| | - Antonio Vendramin
- Department of Oncology and Hemato-Oncology; University of Milan; Milan Italy
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
| | - Chiara De Philippis
- Department of Oncology and Hemato-Oncology; University of Milan; Milan Italy
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
| | - Nikhil C. Munshi
- LeBow Institute for Myeloma Therapeutics; Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School; Boston Massachusetts
| | - Herve Avet-Loiseau
- Laboratory for Genomics in Myeloma; University Cancer Center of Toulouse; CRCT INSERM 1037, Toulouse France
| | - Peter J. Campbell
- The Cancer Genome Project; Wellcome Trust Sanger Institute; Cambridge United Kingdom
| | - Alberto Mussetti
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
| | - Cristiana Carniti
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
| | - Francesco Maura
- Department of Oncology and Hemato-Oncology; University of Milan; Milan Italy
| | - Cristina Barlassina
- Genomic and Bioinformatics Unit; Department of Health Sciences, University of Milan; Milan Italy
| | - Paolo Corradini
- Department of Oncology and Hemato-Oncology; University of Milan; Milan Italy
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
| | - Vittorio Montefusco
- Department of Hematology and Pediatric Onco-Hematology; IRCCS Foundation National Cancer Institute; Milan Italy
| |
Collapse
|
390
|
Xu J, Pfarr N, Endris V, Mai EK, Md Hanafiah NH, Lehners N, Penzel R, Weichert W, Ho AD, Schirmacher P, Goldschmidt H, Andrulis M, Raab MS. Molecular signaling in multiple myeloma: association of RAS/RAF mutations and MEK/ERK pathway activation. Oncogenesis 2017; 6:e337. [PMID: 28504689 PMCID: PMC5523069 DOI: 10.1038/oncsis.2017.36] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 02/10/2017] [Accepted: 03/28/2017] [Indexed: 12/27/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy that is still considered to be incurable in most cases. A dominant mutation cluster has been identified in RAS/RAF genes, emphasizing the potential significance of RAS/RAF/MEK/ERK signaling as a therapeutic target. As yet, however, the clinical relevance of this finding is unclear as clinical responses to MEK inhibition in RAS-mutant MM have been mixed. We therefore assessed RAS/RAF mutation status and MEK/ERK pathway activation by both targeted sequencing and phospho-ERK immunohistochemistry in 180 tissue biopsies from 103 patients with newly diagnosed MM (NDMM) and 77 patients with relapsed/refractory MM (rrMM). We found a significant enrichment of RAS/BRAF mutations in rrMM compared to NDMM (P=0.011), which was mainly due to an increase of NRAS mutations (P=0.010). As expected, BRAF mutations were significantly associated with activated downstream signaling. However, only KRAS and not NRAS mutations were associated with pathway activation compared to RAS/BRAFwt (P=0.030). More specifically, only KRASG12D and BRAFV600E were consistently associated with ERK activation (P<0.001 and P=0.006, respectively). Taken together, these results suggest the need for a more specific stratification strategy consisting of both confirmation of protein-level pathway activation as well as detailed RAS/RAF mutation status to allow for a more precise and more effective application of targeted therapies, for example, with BRAF/MEK inhibitors in MM.
Collapse
Affiliation(s)
- J Xu
- Max Eder Group Experimental Therapies for Hematologic Malignancies, Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - N Pfarr
- Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - V Endris
- Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - E K Mai
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - N H Md Hanafiah
- Max Eder Group Experimental Therapies for Hematologic Malignancies, Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - N Lehners
- Max Eder Group Experimental Therapies for Hematologic Malignancies, Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - R Penzel
- Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - W Weichert
- Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - A D Ho
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - P Schirmacher
- Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - H Goldschmidt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - M Andrulis
- Department of General Pathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - M S Raab
- Max Eder Group Experimental Therapies for Hematologic Malignancies, Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
391
|
KDM6B modulates MAPK pathway mediating multiple myeloma cell growth and survival. Leukemia 2017; 31:2661-2669. [PMID: 28487543 PMCID: PMC5681448 DOI: 10.1038/leu.2017.141] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/11/2017] [Accepted: 05/01/2017] [Indexed: 12/18/2022]
Abstract
Recent studies have delineated cancer-type-specific roles of histone 3 lysine 27 (H3K27) demethylase KDM6B/JMJD3 depending on its H3K27 demethylase activity. Here we show that KDM6B is expressed in multiple myeloma (MM) cells; and that shRNA-mediated knockdown and CRISPR-mediated knockout of KDM6B abrogate MM cell growth and survival. Tumor necrosis factor-α or bone marrow stromal cell culture supernatants induce KDM6B, which is blocked by IKKβ inhibitor MLN120B, suggesting that KDM6B is regulated by NF-κB signaling in MM cells. RNA-seq and subsequent ChIP-qPCR analyses reveal that KDM6B is recruited to the loci of genes encoding components of MAPK signaling pathway including ELK1 and FOS, and upregulates expression of these genes without affecting H3K27 methylation level. Overexpression of catalytically inactive KDM6B activates expression of MAPK pathway-related genes, confirming its function independent of demethylase activity. We further demonstrate that downstream targets of KDM6B, ELK1 and FOS, confer MM cell growth. Our study therefore delineates KDM6B function that links NF-κB and MAPK signaling pathway mediating MM cell growth and survival, and validates KDM6B as a novel therapeutic target in MM.
Collapse
|
392
|
Dutta AK, Hewett DR, Fink JL, Grady JP, Zannettino ACW. Cutting edge genomics reveal new insights into tumour development, disease progression and therapeutic impacts in multiple myeloma. Br J Haematol 2017; 178:196-208. [PMID: 28466550 DOI: 10.1111/bjh.14649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/03/2017] [Accepted: 01/06/2017] [Indexed: 12/19/2022]
Abstract
Multiple Myeloma (MM) is a haematological malignancy characterised by the clonal expansion of plasma cells (PCs) within the bone marrow. Despite advances in therapy, MM remains a largely incurable disease with a median survival of 6 years. In almost all cases, the development of MM is preceded by the benign PC condition Monoclonal Gammopathy of Undetermined Significance (MGUS). Recent studies show that the transformation of MGUS to MM is associated with complex genetic changes. Understanding how these changes contribute to evolution will present targets for clinical intervention. We discuss three models of MM evolution; the linear, the expansionist and the intraclonal heterogeneity models. Of particular interest is the intraclonal heterogeneity model. Here, distinct populations of MM PCs carry differing combinations of genetic mutations. Acquisition of additional mutations can contribute to subclonal lineages where "driver" mutations may influence selective pressure and dominance, and "passenger" mutations are neutral in their effects. Furthermore, studies show that clinical intervention introduces additional selective pressure on tumour cells and can influence subclone survival, leading to therapy resistance. This review discusses how Next Generation Sequencing approaches are revealing critical insights into the genetics of MM development, disease progression and treatment. MM disease progression will illuminate possible mechanisms underlying the tumour.
Collapse
Affiliation(s)
- Ankit K Dutta
- School of Medicine Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Duncan R Hewett
- School of Medicine Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - J Lynn Fink
- The University of Queensland, Diamantina Institute, Woolloongabba, QLD, Australia
| | - John P Grady
- The University of Queensland, Diamantina Institute, Woolloongabba, QLD, Australia
| | - Andrew C W Zannettino
- School of Medicine Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Cancer Theme, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
393
|
Stein CK, Pawlyn C, Chavan S, Rasche L, Weinhold N, Corken A, Buros A, Sonneveld P, Jackson GH, Landgren O, Mughal T, He J, Barlogie B, Bergsagel PL, Davies FE, Walker BA, Morgan GJ. The varied distribution and impact of RAS codon and other key DNA alterations across the translocation cyclin D subgroups in multiple myeloma. Oncotarget 2017; 8:27854-27867. [PMID: 28427158 PMCID: PMC5438613 DOI: 10.18632/oncotarget.15718] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/15/2017] [Indexed: 11/25/2022] Open
Abstract
We examined a set of 805 cases that underwent DNA sequencing using the FoundationOne Heme (F1H) targeted sequencing panel and gene expression profiling. Known and likely variant calls from the mutational data were analyzed for significant associations with gene expression defined translocation cyclin D (TC) molecular subgroups. The spectrum of KRAS, NRAS, and BRAF codon mutations varied across subgroups with NRAS mutations at Q61 codon being common in hyperdiploid (HRD) and t(11;14) myeloma while being rare in MMSET and MAF. In addition, the presence of RAS-RAF mutations was inversely associated with NFκB pathway activation in all subgroups excluding MAF. In the MMSET subgroup, cases with low FGFR3 expression frequently had RAS-RAF mutations. Conditional inference tree analysis determined that mutation and homozygous deletion of TP53, CDKN2C, and RB1 were key prognostic factors associated with adverse outcome in a non-relapse clinical setting. In conclusion, this study highlights the heterogeneity in the distribution and clinical outcomes of RAS codon and other mutations in multiple myeloma dependent upon primary molecular subgroup.
Collapse
Affiliation(s)
- Caleb K. Stein
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Shweta Chavan
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Leo Rasche
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Niels Weinhold
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Adam Corken
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amy Buros
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Graham H. Jackson
- Department of Haematology, Newcastle University, Newcastle-upon-Tyne, UK
| | - Ola Landgren
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tariq Mughal
- FoundationOne Medicine, Cambridge, Massachusetts, USA
- Tufts Medical Center, Boston, Massachusetts, USA
| | - Jie He
- FoundationOne Medicine, Cambridge, Massachusetts, USA
| | - Bart Barlogie
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Faith E. Davies
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Brian A. Walker
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gareth J. Morgan
- The Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
394
|
Hofman IJF, Patchett S, van Duin M, Geerdens E, Verbeeck J, Michaux L, Delforge M, Sonneveld P, Johnson AW, De Keersmaecker K. Low frequency mutations in ribosomal proteins RPL10 and RPL5 in multiple myeloma. Haematologica 2017; 102:e317-e320. [PMID: 28428269 DOI: 10.3324/haematol.2016.162198] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Isabel J F Hofman
- Department of Oncology, KU Leuven - University of Leuven, LKI - Leuven Cancer Institute, Belgium
| | - Stephanie Patchett
- Department of Molecular Biosciences, The University of Texas at Austin, TX, USA
| | - Mark van Duin
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Ellen Geerdens
- Center for Cancer Biology, VIB, Leuven, Belgium.,Center for Human Genetics, KU Leuven - University of Leuven, LKI - Leuven Cancer Institute, Belgium
| | - Jelle Verbeeck
- Department of Oncology, KU Leuven - University of Leuven, LKI - Leuven Cancer Institute, Belgium
| | - Lucienne Michaux
- Center for Human Genetics, KU Leuven - University Hospitals Leuven, Belgium
| | - Michel Delforge
- LKI - Leuven Cancer Institute, Department of Development and Regeneration, KU Leuven - University Hospitals Leuven, Belgium
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Arlen W Johnson
- Department of Molecular Biosciences, The University of Texas at Austin, TX, USA
| | - Kim De Keersmaecker
- Department of Oncology, KU Leuven - University of Leuven, LKI - Leuven Cancer Institute, Belgium
| |
Collapse
|
395
|
Rasche L, Weinhold N, Morgan GJ, van Rhee F, Davies FE. Immunologic approaches for the treatment of multiple myeloma. Cancer Treat Rev 2017; 55:190-199. [PMID: 28431262 DOI: 10.1016/j.ctrv.2017.03.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/11/2022]
Abstract
The FDA approval of two monoclonal antibodies in 2015has heralded a new era of targeted immunotherapies for multiple myeloma (MM). In this review we discuss the recent approaches using different immunological components to treat MM. In particular, we review current monoclonal antibody based therapies, engineered T- and NK cell products, 'off-target' immunomodulation, and strategies utilizing allogeneic cell transplantation in MM. We discuss how an immunologic approach offers promise for the treatment of this genetically heterogeneous disease, and how patients with acquired drug resistance may particularly benefit from these therapies. We also describe some of the limitations of the current strategies and speculate on the future of personalized immunotherapies for MM.
Collapse
Affiliation(s)
- Leo Rasche
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Frits van Rhee
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Faith E Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
396
|
Rustad EH, Coward E, Skytøen ER, Misund K, Holien T, Standal T, Børset M, Beisvag V, Myklebost O, Meza-Zepeda LA, Dai HY, Sundan A, Waage A. Monitoring multiple myeloma by quantification of recurrent mutations in serum. Haematologica 2017; 102:1266-1272. [PMID: 28385781 PMCID: PMC5566041 DOI: 10.3324/haematol.2016.160564] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/31/2017] [Indexed: 01/28/2023] Open
Abstract
Circulating tumor DNA is a promising biomarker to monitor tumor load and genome alterations. We explored the presence of circulating tumor DNA in multiple myeloma patients and its relation to disease activity during long-term follow-up. We used digital droplet polymerase chain reaction analysis to monitor recurrent mutations, mainly in mitogen activated protein kinase pathway genes NRAS, KRAS and BRAF. Mutations were identified by next-generation sequencing or polymerase chain reaction analysis of bone marrow plasma cells, and their presence analyzed in 251 archived serum samples obtained from 20 patients during a period of up to 7 years. In 17 of 18 patients, mutations identified in bone marrow during active disease were also found in a time-matched serum sample. The concentration of mutated alleles in serum correlated with the fraction in bone marrow plasma cells (r=0.507, n=34, P<0.002). There was a striking covariation between circulating mutation levels and M protein in ten out of 11 patients with sequential samples. When relapse evaluation by circulating tumor DNA and M protein could be directly compared, the circulating tumor DNA showed relapse earlier in two patients (3 and 9 months), later in one patient (4 months) and in three patients there was no difference. In three patients with transformation to aggressive disease, the concentrations of mutations in serum increased up to 400 times, an increase that was not seen for the M protein. In conclusion, circulating tumor DNA in myeloma is a multi-faceted biomarker reflecting mutated cells, total tumor mass and transformation to a more aggressive disease. Its properties are both similar and complementary to M protein.
Collapse
Affiliation(s)
- Even Holth Rustad
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Eivind Coward
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Norwegian Cancer Genomics Consortium, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Emilie R Skytøen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Kristine Misund
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Toril Holien
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Therese Standal
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,CEMIR - Center for Molecular Inflammation Research, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Magne Børset
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Vidar Beisvag
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Ola Myklebost
- Norwegian Cancer Genomics Consortium, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Institute for Clinical Science, University of Bergen, Trondheim, Norway.,Institute for Cancer Research, Oslo University Hospital, Trondheim, Norway
| | - Leonardo A Meza-Zepeda
- Norwegian Cancer Genomics Consortium, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Institute for Cancer Research, Oslo University Hospital, Trondheim, Norway
| | - Hong Yan Dai
- Department of Pathology and Medical Genetics, St. Olav's University Hospital, Trondheim, Norway
| | - Anders Sundan
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,CEMIR - Center for Molecular Inflammation Research, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Anders Waage
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NTNU, Trondheim, Norway .,Norwegian Cancer Genomics Consortium, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.,Department of Hematology, St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
397
|
Overexpression of EZH2 in multiple myeloma is associated with poor prognosis and dysregulation of cell cycle control. Blood Cancer J 2017; 7:e549. [PMID: 28362441 PMCID: PMC5380911 DOI: 10.1038/bcj.2017.27] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 02/23/2017] [Indexed: 12/31/2022] Open
Abstract
Myeloma is heterogeneous at the molecular level with subgroups of patients characterised by features of epigenetic dysregulation. Outcomes for myeloma patients have improved over the past few decades except for molecularly defined high-risk patients who continue to do badly. Novel therapeutic approaches are, therefore, required. A growing number of epigenetic inhibitors are now available including EZH2 inhibitors that are in early-stage clinical trials for treatment of haematological and other cancers with EZH2 mutations or in which overexpression has been correlated with poor outcomes. For the first time, we have identified and validated a robust and independent deleterious effect of high EZH2 expression on outcomes in myeloma patients. Using two chemically distinct small-molecule inhibitors, we demonstrate a reduction in myeloma cell proliferation with EZH2 inhibition, which leads to cell cycle arrest followed by apoptosis. This is mediated via upregulation of cyclin-dependent kinase inhibitors associated with removal of the inhibitory H3K27me3 mark at their gene loci. Our results suggest that EZH2 inhibition may be a potential therapeutic strategy for the treatment of myeloma and should be investigated in clinical studies.
Collapse
|
398
|
Abstract
Personalization of therapy to target specific molecular pathways has been placed in the forefront of cancer research. Initial reports from clinical trials designed to select patients for appropriate treatment on the basis of tumor characteristics not only have generated considerable excitement but also have identified several challenges. These challenges include the overcoming of regulatory and logistic difficulties, identification of the best selection biomarkers and diagnostic platforms that can be applied in the clinical setting, definition of relevant outcomes in small preselected patient populations, and the design of methods that facilitate rapid enrollment and interpretation of clinical trials by aggregating data across histologically diverse malignancies with common genetic alterations. Furthermore, because our knowledge of the functional consequences of many genetic alterations lags, investigators and sponsors struggle with choosing between ideal clinical trial designs and more practical ones. These challenges are amplified when more than one biomarker is used to select patients for a combination of targeted agents. This review summarizes the current status and challenges of clinical trials in the genomic era and proposes ways to address these challenges.
Collapse
Affiliation(s)
- Erel Joffe
- All authors: Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexia Iasonos
- All authors: Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anas Younes
- All authors: Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
399
|
Robiou du Pont S, Cleynen A, Fontan C, Attal M, Munshi N, Corre J, Avet-Loiseau H. Genomics of Multiple Myeloma. J Clin Oncol 2017; 35:963-967. [DOI: 10.1200/jco.2016.70.6705] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is characterized by wide variability in the chromosomal/genetic changes present in tumor plasma cells. Genetically, MM can be divided into two groups according to ploidy and hyperdiploidy versus nonhyperdiploidy. Several studies in gene expression profiling attempted to identify subentities in MM without convincing results. These studies mostly confirmed the cytogenetic data and subclassified patients according to 14q32 translocations and ploidy. More-recent data that are based on whole-exome sequencing have confirmed this heterogeneity and show many gene mutations but without a unifying mutation. These newer studies have shown the frequent alteration of the mitogen-activated protein kinase pathway. The most interesting data have demonstrated subclonality in all patients with MM, including subclonal mutations of supposed driver genes KRAS, NRAS, and BRAF.
Collapse
Affiliation(s)
- Sebastien Robiou du Pont
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| | - Alice Cleynen
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| | - Charlotte Fontan
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| | - Michel Attal
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| | - Nikhil Munshi
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| | - Jill Corre
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| | - Hervé Avet-Loiseau
- Sebastien Robiou du Pont, Charlotte Fontan, Michel Attal, Jill Corre, and Hervé Avet-Loiseau, L’Institut Universitaire du Cancer Oncopole, Toulouse; Alice Cleynen, Centre National de la Recherche Scientifique, and Montpellier University, Montpellier, France; and Nikhil Munshi, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
400
|
Tessoulin B, Eveillard M, Lok A, Chiron D, Moreau P, Amiot M, Moreau-Aubry A, Le Gouill S, Pellat-Deceunynck C. p53 dysregulation in B-cell malignancies: More than a single gene in the pathway to hell. Blood Rev 2017; 31:251-259. [PMID: 28284458 DOI: 10.1016/j.blre.2017.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 03/02/2017] [Indexed: 12/31/2022]
Abstract
TP53 deletion or mutation is frequent in B-cell malignancies and is associated with a low response rate. We describe here the p53 landscape in B-cell malignancies, from B-Acute Lymphoblastic Leukemia to Plasma Cell Leukemia, by analyzing incidence of gain or loss of function of actors both upstream and within the p53 pathway, namely MYC, RAS, ARF, MDM2, ATM and TP53. Abnormalities are not equally distributed and their incidence is highly variable among malignancies. Deletion and mutation, usually associated, of ATM or TP53 are frequent in Diffuse Large B-Cell Lymphoma and Mantle Cell Lymphoma. MYC gain, absent in post-GC malignancies, is frequent in B-Prolymphocytic-Leukemia, Multiple Myeloma and Plasma Cell Leukemias. RAS mutations are rare except in MM and PCL. Multiple Factorial Analysis notes that MYC deregulation is closely related to TP53 status. Moreover, MYC gain, TP53 deletion and RAS mutations are inversely correlated with survival. Based on this landscape, we further propose targeted therapeutic approaches for the different B-cell malignancies.
Collapse
Affiliation(s)
- B Tessoulin
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France.
| | - M Eveillard
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Hematology Biology Department, Nantes University Hospital, Nantes, France
| | - A Lok
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France
| | - D Chiron
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - P Moreau
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France
| | - M Amiot
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - A Moreau-Aubry
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France
| | - S Le Gouill
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France; Department of Hematology, Nantes University Hospital, Nantes, France
| | - C Pellat-Deceunynck
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, France.
| |
Collapse
|