301
|
Pugh TJ, Fink JM, Lu X, Mathew S, Murata-Collins J, Willem P, Fang M. Assessing genome-wide copy number aberrations and copy-neutral loss-of-heterozygosity as best practice: An evidence-based review from the Cancer Genomics Consortium working group for plasma cell disorders. Cancer Genet 2018; 228-229:184-196. [DOI: 10.1016/j.cancergen.2018.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 12/28/2022]
|
302
|
The genomic landscape of plasma cells in systemic light chain amyloidosis. Blood 2018; 132:2775-2777. [PMID: 30446495 DOI: 10.1182/blood-2018-08-872226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/06/2018] [Indexed: 11/20/2022] Open
|
303
|
Chapman MA, Sive J, Ambrose J, Roddie C, Counsell N, Lach A, Abbasian M, Popat R, Cavenagh JD, Oakervee H, Streetly MJ, Schey S, Koh M, Willis F, Virchis AE, Crowe J, Quinn MF, Cook G, Crawley CR, Pratt G, Cook M, Braganza N, Adedayo T, Smith P, Clifton-Hadley L, Owen RG, Sonneveld P, Keats JJ, Herrero J, Yong K. RNA-seq of newly diagnosed patients in the PADIMAC study leads to a bortezomib/lenalidomide decision signature. Blood 2018; 132:2154-2165. [PMID: 30181174 PMCID: PMC6310235 DOI: 10.1182/blood-2018-05-849893] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/28/2018] [Indexed: 01/29/2023] Open
Abstract
Improving outcomes in multiple myeloma will involve not only development of new therapies but also better use of existing treatments. We performed RNA sequencing on samples from newly diagnosed patients enrolled in the phase 2 PADIMAC (Bortezomib, Adriamycin, and Dexamethasone Therapy for Previously Untreated Patients with Multiple Myeloma: Impact of Minimal Residual Disease in Patients with Deferred ASCT) study. Using synthetic annealing and the large margin nearest neighbor algorithm, we developed and trained a 7-gene signature to predict treatment outcome. We tested the signature in independent cohorts treated with bortezomib- and lenalidomide-based therapies. The signature was capable of distinguishing which patients would respond better to which regimen. In the CoMMpass data set, patients who were treated correctly according to the signature had a better progression-free survival (median, 20.1 months vs not reached; hazard ratio [HR], 0.40; confidence interval [CI], 0.23-0.72; P = .0012) and overall survival (median, 30.7 months vs not reached; HR, 0.41; CI, 0.21-0.80; P = .0049) than those who were not. Indeed, the outcome for these correctly treated patients was noninferior to that for those treated with combined bortezomib, lenalidomide, and dexamethasone, arguably the standard of care in the United States but not widely available elsewhere. The small size of the signature will facilitate clinical translation, thus enabling more targeted drug regimens to be delivered in myeloma.
Collapse
Affiliation(s)
- Michael A Chapman
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Jonathan Sive
- Department of Haemato-oncology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - John Ambrose
- Bill Lyons Informatics Centre, UCL Cancer Institute, University College London, London, United Kingdom
| | - Claire Roddie
- Department of Haematology, University College London Hospitals, London, United Kingdom
| | - Nicholas Counsell
- Cancer Research UK and UCL Cancer Trials Centre, London, United Kingdom
| | - Anna Lach
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Mahnaz Abbasian
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Rakesh Popat
- Department of Haematology, University College London Hospitals, London, United Kingdom
| | - Jamie D Cavenagh
- Department of Haemato-oncology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Heather Oakervee
- Department of Haemato-oncology, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Matthew J Streetly
- Department of Haematology, Guys and St. Thomas' Hospital, London, United Kingdom
| | - Stephen Schey
- Department of Haematology, Kings College Hospital, London, United Kingdom
| | - Mickey Koh
- Department of Haematology, St. George's Hospital, London, United Kingdom
| | - Fenella Willis
- Department of Haematology, St. George's Hospital, London, United Kingdom
| | - Andres E Virchis
- Department of Haematology, Royal Free London, Barnet and Chase Farm Hospitals, London, United Kingdom
| | - Josephine Crowe
- Department of Haematology, Royal United Hospitals Bath, Bath, United Kingdom
| | - Michael F Quinn
- Department of Haematology, Belfast City Hospital, Belfast, United Kingdom
| | - Gordon Cook
- Department of Haematology, St. James's University Hospital, Leeds, United Kingdom
| | - Charles R Crawley
- Department of Haematology, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Guy Pratt
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Mark Cook
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, United Kingdom
| | - Nivette Braganza
- Cancer Research UK and UCL Cancer Trials Centre, London, United Kingdom
| | - Toyin Adedayo
- Cancer Research UK and UCL Cancer Trials Centre, London, United Kingdom
| | - Paul Smith
- Cancer Research UK and UCL Cancer Trials Centre, London, United Kingdom
| | | | - Roger G Owen
- Haematological Malignancy Diagnostic Service, St. James' University Hospital, Leeds, United Kingdom
| | | | - Jonathan J Keats
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, AZ
| | - Javier Herrero
- Bill Lyons Informatics Centre, UCL Cancer Institute, University College London, London, United Kingdom
| | - Kwee Yong
- Department of Haematology, UCL Cancer Institute, University College London, London, United Kingdom
| |
Collapse
|
304
|
Perrot A, Corre J, Avet-Loiseau H. Risk Stratification and Targets in Multiple Myeloma: From Genomics to the Bedside. Am Soc Clin Oncol Educ Book 2018; 38:675-680. [PMID: 30231368 DOI: 10.1200/edbk_200879] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the past 15 years, significant improvements in overall survival have been observed in multiple myeloma (MM), mainly due to the availability of novel drugs with variable mechanisms of action. However, these improvements do not benefit all patients, and some of them, defined as high risk, still display short survival. The most important risk factors are the genetic abnormalities present in the malignant plasma cells. The most important high-risk features are the del(17p), the del(1p32), the t(4;14), and 1q gains. Assessing these markers is mandatory at diagnosis and at least at first relapse, since it has been clearly shown that the lenalidomide-dexamethasone combination is not efficient in these high-risk patients. In contrast, a triplet combination adding a proteasome inhibitor or a monoclonal antibody to the lenalidomide-dexamethasone backbone clearly improves the survival. Another way to improve the outcome would be to specifically target genetic abnormalities with specific inhibitors. The sequencing of more than 1,000 MM exomes revealed again a huge heterogeneity. The most frequent mutations involve the KRAS and NRAS genes (20%-25% each). However, to date, no good RAS-inhibitors are clinically available, preventing targeted therapy. The only drugable target is the V600E BRAF mutation. Unfortunately, this specific mutation is present in only 3% of the patients. Finally, it has been recently reported a specific efficiency of the BCL2-inhibitor venetoclax in patients with the t(11;14) translocation, which is found in 20% of the patients.
Collapse
Affiliation(s)
- Aurore Perrot
- From the Hematology Department, University Hospital, Nancy, France; Myeloma Genomics Laboratory, University Hospital, Toulouse, France; and Myeloma Genomics Laboratory, University Hospital, Toulouse, France
| | - Jill Corre
- From the Hematology Department, University Hospital, Nancy, France; Myeloma Genomics Laboratory, University Hospital, Toulouse, France; and Myeloma Genomics Laboratory, University Hospital, Toulouse, France
| | - Hervé Avet-Loiseau
- From the Hematology Department, University Hospital, Nancy, France; Myeloma Genomics Laboratory, University Hospital, Toulouse, France; and Myeloma Genomics Laboratory, University Hospital, Toulouse, France
| |
Collapse
|
305
|
Levin A, Hari P, Dhakal B. Novel biomarkers in multiple myeloma. Transl Res 2018; 201:49-59. [PMID: 30301522 DOI: 10.1016/j.trsl.2018.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/10/2018] [Accepted: 05/22/2018] [Indexed: 11/24/2022]
Abstract
Significant advancements have been made in the molecular mechanisms of myelomagenesis, diagnostic methods, prognostication, and the treatment options in multiple myeloma (MM) over the last decade. Despite these, MM remains a heterogeneous disease with differing outcomes. As myeloma treatment landscape continues to expand, personalized treatment that provides maximum benefit to a specific patient becomes more important. In the last few years, serum monoclonal proteins including the serum-free light chain assays, imaging, and cytogenetics have been used to predict the outcomes of MM patients receiving different types of therapies. With the development of novel technologies, more sensitive detection of residual disease using flow cytometry and next-generation sequencing has been possible. In addition, liquid biopsies using circulating tumor cells, tumor DNA, and novel immune biomarkers are potentially being investigated. These novel potential biomarkers not only accurately detect the mutational landscape of different cancers compared to standard methods but also serve as prognostic and predictive biomarkers for disease relapse and response to therapy. It is likely that we will be able to offer more targeted and risk-adapted therapeutic approach to patients with MM at different stages of their disease guided by these potential biomarkers.
Collapse
Affiliation(s)
- Adam Levin
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Parameswaran Hari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Binod Dhakal
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
306
|
Subclonal TP53 copy number is associated with prognosis in multiple myeloma. Blood 2018; 132:2465-2469. [PMID: 30373884 DOI: 10.1182/blood-2018-06-857250] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/03/2018] [Indexed: 02/03/2023] Open
Abstract
Multiple myeloma (MM) is a genetically heterogeneous cancer of bone marrow plasma cells with variable outcome. To assess the prognostic relevance of clonal heterogeneity of TP53 copy number, we profiled tumors from 1777 newly diagnosed Myeloma XI trial patients with multiplex ligation-dependent probe amplification (MLPA). Subclonal TP53 deletions were independently associated with shorter overall survival, with a hazard ratio of 1.8 (95% confidence interval, 1.2-2.8; P = .01). Clonal, but not subclonal, TP53 deletions were associated with clinical markers of advanced disease, specifically lower platelet counts (P < .001) and increased lactate dehydrogenase (P < .001), as well as a higher frequency of features indicative of genomic instability, del(13q) (P = .002) or del(1p) (P = .006). Biallelic TP53 loss-of-function by mutation and deletion was rare (2.4%) and associated with advanced disease. We present a framework for identifying subclonal TP53 deletions by MLPA, to improve patient stratification in MM and tailor therapy, enabling management strategies.
Collapse
|
307
|
Xia E, Kanematsu S, Suenaga Y, Elzawahry A, Kondo H, Otsuka N, Moriya Y, Iizasa T, Kato M, Yoshino I, Yokoi S. MicroRNA induction by copy number gain is associated with poor outcome in squamous cell carcinoma of the lung. Sci Rep 2018; 8:15363. [PMID: 30337605 PMCID: PMC6194131 DOI: 10.1038/s41598-018-33696-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Copy number gains in cancer genomes have been shown to induce oncogene expression and promote carcinogenesis; however, their role in regulating oncogenic microRNAs (onco-miRNAs) remains largely unknown. Our aim was to identify onco-miRNAs induced by copy number gains in human squamous cell carcinoma (Sq) of the lung. We performed a genome-wide screen of onco-miRNAs from 245 Sqs using data sets from RNA-sequencing, comparative genomic hybridization, and the corresponding clinical information from The Cancer Genome Atlas. Among 1001 miRNAs expressed in the samples, 231 were correlated with copy number alternations, with only 11 of these being highly expressed in Sq compared to adenocarcinoma and normal tissues. Notably, miR-296-5p, miR-324-3p, and miR-3928-3p expression was significantly associated with poor prognosis. Multivariate analysis using the Cox proportional hazards model showed that miRNA expression and smoking were independent prognostic factors and were associated with poor prognosis. Furthermore, the three onco-miRNAs inhibited FAM46C to induce MYC expression, promoting proliferation of Sq cells. We found that copy number gains in Sq of the lung induce onco-miRNA expression that is associated with poor prognosis.
Collapse
Affiliation(s)
- Endi Xia
- Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan.,Department of General Thoracic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sotaro Kanematsu
- Division of Genetic Diagnostics, Chiba Cancer Center, Chiba, Japan
| | - Yusuke Suenaga
- Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Asmaa Elzawahry
- Department of Bioinformatics, National Cancer Center, Tokyo, Japan
| | - Hitomi Kondo
- Division of Genetic Diagnostics, Chiba Cancer Center, Chiba, Japan
| | - Noriko Otsuka
- Division of Genetic Diagnostics, Chiba Cancer Center, Chiba, Japan
| | - Yasumitsu Moriya
- Division of Thoracic Diseases, Chiba Cancer Center, Chiba, Japan
| | - Toshihiko Iizasa
- Division of Thoracic Diseases, Chiba Cancer Center, Chiba, Japan
| | - Mamoru Kato
- Department of Bioinformatics, National Cancer Center, Tokyo, Japan
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sana Yokoi
- Cancer Genome Center, Chiba Cancer Center Research Institute, Chiba, Japan. .,Division of Genetic Diagnostics, Chiba Cancer Center, Chiba, Japan.
| |
Collapse
|
308
|
Maes K, Boeckx B, Vlummens P, De Veirman K, Menu E, Vanderkerken K, Lambrechts D, De Bruyne E. The genetic landscape of 5T models for multiple myeloma. Sci Rep 2018; 8:15030. [PMID: 30301958 PMCID: PMC6177465 DOI: 10.1038/s41598-018-33396-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/25/2018] [Indexed: 12/24/2022] Open
Abstract
Murine models for multiple myeloma (MM) are often used to investigate pathobiology of multiple myeloma and disease progression. Unlike transgenic mice models, where it is known which oncogene is driving MM disease, the somatic aberrations of spontaneous syngeneic 5T models of MM have not yet been reported. Here, we analyzed the copy-number alterations (CNA) and mutational landscape of 5T2, 5T33vv and 5TGM1 murine MM models using whole-genome and whole-exome sequencing. Forty four percent of the genome of 5T2 cells is affected by CNAs while this was only 11% and 17% for 5T33vv and 5TGM1 cells, respectively. We found that up to 69% of the genes linked to gain of 1q or deletion of 13q in MM patients are present as respectively gains in 5T2 cells or deletions in 5T33 and 5TGM1 cells. Exome sequencing furthermore revealed mutations of genes involved in RAS/MAPK, PI3K/AKT1 and JAK/STAT signaling, DNA damage response, cell cycle, epigenetic regulation and extracellular matrix organization. We observed a statistically significant overlap of genes mutated in the 5T models and MM patients. Overall, the genetic landscape of the 5T models is heterogeneous with a high number of aberrations involving genes in various multiple myeloma-related pathways.
Collapse
Affiliation(s)
- Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, 1090, Belgium.
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, Katholieke Universiteit Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
| | - Philip Vlummens
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, 1090, Belgium
- Department of Clinical Hematology, Ghent University Hospital, Gent, 9000, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, 1090, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, 1090, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, 1090, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, Katholieke Universiteit Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Brussel, 1090, Belgium
| |
Collapse
|
309
|
Maffini E, Storer BE, Sandmaier BM, Bruno B, Sahebi F, Shizuru JA, Chauncey TR, Hari P, Lange T, Pulsipher MA, McSweeney PA, Holmberg L, Becker PS, Green DJ, Mielcarek M, Maloney DG, Storb R. Long-term follow up of tandem autologous-allogeneic hematopoietic cell transplantation for multiple myeloma. Haematologica 2018; 104:380-391. [PMID: 30262560 PMCID: PMC6355483 DOI: 10.3324/haematol.2018.200253] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022] Open
Abstract
We previously reported initial results in 102 multiple myeloma (MM) patients treated with sequential high-dose melphalan and autologous hematopoietic cell transplantation followed by 200 cGy total body irradiation with or without fludarabine 90 mg/m2 and allogeneic hematopoietic cell transplantation. Here we present long-term clinical outcomes among the 102 initial patients and among 142 additional patients, with a median follow up of 8.3 (range 1.0-18.1) years. Donors included human leukocyte antigen identical siblings (n=179) and HLA-matched unrelated donors (n=65). A total of 209 patients (86%) received tandem autologous-allogeneic upfront, while thirty-five patients (14%) had failed a previous autologous hematopoietic cell transplantation before the planned autologous-allogeneic transplantation. Thirty-one patients received maintenance treatment at a median of 86 days (range, 61-150) after allogeneic transplantation. Five-year rates of overall survival (OS) and progression-free survival (PFS) were 54% and 31%, respectively. Ten-year OS and PFS were 41% and 19%, respectively. Overall non-relapse mortality was 2% at 100 days and 14% at five years. Patients with induction-refractory disease and those with high-risk biological features experienced shorter OS and PFS. A total of 152 patients experienced disease relapse and 117 of those received salvage treatment. Eighty-three of the 117 patients achieved a clinical response, and for those, the median duration of survival after relapse was 7.8 years. Moreover, a subset of patients who became negative for minimal residual disease (MRD) by flow cytometry experienced a significantly lower relapse rate as compared with MRD-positive patients (P=0.03). Our study showed that the graft-versus-myeloma effect after non-myeloablative allografting allowed long-term disease control in standard and high-risk patient subsets. Ultra-high-risk patients did not appear to benefit from tandem autologous/allogeneic hematopoietic cell transplantation because of early disease relapse. Incorporation of newer anti-MM agents into the initial induction treatments before tandem hematopoietic cell transplantation and during maintenance might improve outcomes of ultra-high-risk patients. Clinical trials included in this study are registered at: clinicaltrials.gov identifiers: 00075478, 00005799, 01251575, 00078858, 00105001, 00027820, 00089011, 00003196, 00006251, 00793572, 00054353, 00014235, 00003954.
Collapse
Affiliation(s)
- Enrico Maffini
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA
| | - Barry E Storer
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,University of Washington School of Public Health, Seattle, WA, USA
| | - Brenda M Sandmaier
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,Department of Medicine, Seattle, WA, USA
| | - Benedetto Bruno
- University of Turin, Department of Molecular Biotechnology and Health Sciences, Turin, Italy
| | - Firoozeh Sahebi
- City of Hope National Medical Center/Southern California Kaiser Permanente Medical Group, Duarte, CA, USA
| | | | - Thomas R Chauncey
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,Department of Medicine, Seattle, WA, USA.,VA Puget Sound Medical Health Care System, Seattle, WA, USA
| | | | | | | | | | - Leona Holmberg
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,University of Washington School of Public Health, Seattle, WA, USA
| | - Pamela S Becker
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,Department of Medicine, Seattle, WA, USA
| | - Damian J Green
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,Department of Medicine, Seattle, WA, USA
| | - Marco Mielcarek
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,Department of Medicine, Seattle, WA, USA
| | - David G Maloney
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA.,Department of Medicine, Seattle, WA, USA
| | - Rainer Storb
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA, USA .,Department of Medicine, Seattle, WA, USA
| |
Collapse
|
310
|
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; 9:2111. [PMID: 30294322 PMCID: PMC6158389 DOI: 10.3389/fimmu.2018.02111] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/28/2018] [Indexed: 12/25/2022] 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
|
311
|
Wong KY, Yao Q, Yuan LQ, Li Z, Ma ESK, Chim CS. Frequent functional activation of RAS signalling not explained by RAS/RAF mutations in relapsed/refractory multiple myeloma. Sci Rep 2018; 8:13522. [PMID: 30201956 PMCID: PMC6131153 DOI: 10.1038/s41598-018-31820-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
RAS mutations are frequent in relapsed/refractory multiple myeloma (RRMM) but functional study in primary samples is scanty. Herein, in primary myeloma plasma cells of 17 suspected RRMM, functional activation of RAS signalling was studied by Western blot of phosphorylated ERK1/2 (phospho-ERK1/2). Moreover, activating mutations in KRAS, NRAS, BRAF, and ALK were studied by PCR and bidirectional direct sequencing. Furthermore, methylation of negative RAS signalling regulator genes, RASSF1A and RASD1, were analyzed by methylation-specific PCR. As evidenced by phospho-ERK1/2 over-expression, functional RAS activation was detected in 12 (75.0%) RRMM. Of patients with functional RAS activation, sequencing data showed only seven (58.3%) patients with one each had NRAS Q61H, NRAS Q61K, KRAS G12D, KRAS G12V, KRAS G13D, KRAS Q61P, or BRAF V600E mutation, whereas five (41.7%) patients had no RAS/RAF mutation. Conversely, patients without functional RAS activation had no RAS/RAF mutation. Moreover, none of the patients with functional RAS activation had ALK mutations, or methylation of RASSF1A and RASD1. Collectively, functional activation of RAS signalling was present in majority of RRMM but only about half (58.3%) accountable by RAS/RAF mutations. If verified in larger studies, clinical investigations of MEK inhibitors are warranted regardless of RAS/RAF mutations.
Collapse
Affiliation(s)
- Kwan Yeung Wong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Qiumei Yao
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenhai Li
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong
| | - Edmond Shiu Kwan Ma
- Department of Pathology, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam Road, Pokfulam, Hong Kong.
| |
Collapse
|
312
|
Abstract
The enhancer of zeste homolog 2 (EZH2) is the enzymatic subunit of the polycomb repressive complex 2 (PRC2) that exerts important functions during normal development as well as disease. PRC2 through EZH2 tri-methylates histone H3 lysine tail residue 27 (H3K27me3), a modification associated with repression of gene expression programs related to stem cell self-renewal, cell cycle, cell differentiation, and cellular transformation. EZH2 is deregulated and subjected to gain of function or loss of function mutations, and hence functions as an oncogene or tumor suppressor gene in a context-dependent manner. The development of highly selective inhibitors against the histone methyltransferase activity of EZH2 has also contributed to insight into the role of EZH2 and PRC2 in tumorigenesis, and their potential as therapeutic targets in cancer. EZH2 can function as an oncogene in multiple myeloma (MM) by repressing tumor suppressor genes that control apoptosis, cell cycle control and adhesion properties. Taken together these findings have raised the possibility that EZH2 inhibitors could be a useful therapeutic modality in MM alone or in combination with other targeted agents in MM. Therefore, we review the current knowledge on the regulation of EZH2 and its biological impact in MM, the anti-myeloma activity of EZH2 inhibitors and their potential as a targeted therapy in MM.
Collapse
Affiliation(s)
- Mohammad Alzrigat
- Division of Hematology and Oncology, Department of Medicine, University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA;
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, SE-75185 Uppsala, Sweden;
| | - Jonathan D Licht
- Division of Hematology and Oncology, Department of Medicine, University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA;
| |
Collapse
|
313
|
Raje N, Chau I, Hyman DM, Ribrag V, Blay JY, Tabernero J, Elez E, Wolf J, Yee AJ, Kaiser M, Landau H, Michot JM, Hollebecque A, Veronese L, Makrutzki M, Pitcher B, Puzanov I, Baselga J. Vemurafenib in Patients With Relapsed Refractory Multiple Myeloma Harboring BRAF V600 Mutations: A Cohort of the Histology-Independent VE-BASKET Study. JCO Precis Oncol 2018; 2:1800070. [PMID: 32913989 DOI: 10.1200/po.18.00070] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Noopur Raje
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Ian Chau
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - David M Hyman
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Vincent Ribrag
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Jean-Yves Blay
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Josep Tabernero
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Elena Elez
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Jürgen Wolf
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Andrew J Yee
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Martin Kaiser
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Heather Landau
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Jean-Marie Michot
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Antoine Hollebecque
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Luisa Veronese
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Martina Makrutzki
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Bethany Pitcher
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Igor Puzanov
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| | - Jose Baselga
- and Massachusetts General Hospital, Boston, MA; and Royal Marsden Hospital, Sutton, Surrey, United Kingdom; and Memorial Sloan Kettering Cancer Center, New York; Roswell Park Cancer Institute, Buffalo, NY; and Institut Gustave Roussy, Villejuif; Centre Leon-Berard, Lyon, France; and , Vall d'Hebron University Hospital and Institute of Oncology, Universitat Autònoma de Barcelona, Barcelona, Spain; University Hospital Köln, Köln, Germany; and F. Hoffmann-La Roche Ltd, Basel, Switzerland; and F. Hoffmann-La Roche Ltd, Mississauga, Ontario, Canada
| |
Collapse
|
314
|
Bolli N, Maura F, Minvielle S, Gloznik D, Szalat R, Fullam A, Martincorena I, Dawson KJ, Samur MK, Zamora J, Tarpey P, Davies H, Fulciniti M, Shammas MA, Tai YT, Magrangeas F, Moreau P, Corradini P, Anderson K, Alexandrov L, Wedge DC, Avet-Loiseau H, Campbell P, Munshi N. Genomic patterns of progression in smoldering multiple myeloma. Nat Commun 2018; 9:3363. [PMID: 30135448 PMCID: PMC6105687 DOI: 10.1038/s41467-018-05058-y] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 06/06/2018] [Indexed: 12/21/2022] Open
Abstract
We analyzed whole genomes of unique paired samples from smoldering multiple myeloma (SMM) patients progressing to multiple myeloma (MM). We report that the genomic landscape, including mutational profile and structural rearrangements at the smoldering stage is very similar to MM. Paired sample analysis shows two different patterns of progression: a "static progression model", where the subclonal architecture is retained as the disease progressed to MM suggesting that progression solely reflects the time needed to accumulate a sufficient disease burden; and a "spontaneous evolution model", where a change in the subclonal composition is observed. We also observe that activation-induced cytidine deaminase plays a major role in shaping the mutational landscape of early subclinical phases, while progression is driven by APOBEC cytidine deaminases. These results provide a unique insight into myelomagenesis with potential implications for the definition of smoldering disease and timing of treatment initiation.
Collapse
Affiliation(s)
- Niccolò Bolli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20122, Italy
- Department of Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Francesco Maura
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20122, Italy
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Stephane Minvielle
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, 44035, France
- CHU de Nantes, Nantes, 44093, France
| | - Dominik Gloznik
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Raphael Szalat
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Anthony Fullam
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Inigo Martincorena
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Kevin J Dawson
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Mehmet Kemal Samur
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Jorge Zamora
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Patrick Tarpey
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Helen Davies
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Mariateresa Fulciniti
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Masood A Shammas
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Yu Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Florence Magrangeas
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, 44035, France
- CHU de Nantes, Nantes, 44093, France
| | - Philippe Moreau
- CRCINA, INSERM, CNRS, Université de Nantes, Université d'Angers, Nantes, 44035, France
- CHU de Nantes, Nantes, 44093, France
| | - Paolo Corradini
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20122, Italy
- Department of Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Kenneth Anderson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA
| | - Ludmil Alexandrov
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - David C Wedge
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Herve Avet-Loiseau
- Genomics of Myeloma Laboratory, L'Institut Universitaire du Cancer Oncopole, Toulouse, 31100, France.
| | - Peter Campbell
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, 20122, Italy.
| | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, 02215, MA, USA.
- Veterans Administration Boston Healthcare System, West Roxbury, 02132, MA, USA.
| |
Collapse
|
315
|
Kosztolányi S, Kiss R, Atanesyan L, Gángó A, de Groot K, Steenkamer M, Jáksó P, Matolcsy A, Kajtár B, Pajor L, Szuhai K, Savola S, Bödör C, Alpár D. High-Throughput Copy Number Profiling by Digital Multiplex Ligation-Dependent Probe Amplification in Multiple Myeloma. J Mol Diagn 2018; 20:777-788. [PMID: 30096382 DOI: 10.1016/j.jmoldx.2018.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/09/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a genetically heterogeneous disease with a diverse clinical outcome. Copy number alterations (CNAs), including whole chromosome and subchromosomal gains and losses, are common contributors of the pathogenesis and have demonstrated prognostic impact in MM. We tested the performance of digital multiplex ligation-dependent probe amplification (digitalMLPA), a novel technique combining MLPA and next-generation sequencing, to detect disease-related CNAs. Copy number status at 371 genomic loci was simultaneously analyzed in 56 diagnostic bone marrow samples, which were also examined by conventional MLPA and interphase fluorescence in situ hybridization (iFISH). On average, digitalMLPA identified 4.4 subchromosomal CNAs per patient. The increased number of probes compared with conventional MLPA allowed a detailed mapping of CNAs, especially on chromosome 1, where 24 different patterns were observed in 38 patients harboring loss(1p) and/or gain(1q). iFISH, MLPA, and digitalMLPA results at loci investigated by multiple methods showed a congruency of 95%. Besides precise characterization of hyperdiploid karyotypes not efficiently achievable by iFISH or MLPA, digitalMLPA unraveled 156 CNAs not detected by the other two methods in 45 patients (80%). In addition, we provide proof of principle that digitalMLPA can detect known point mutations, in this case the BRAFV600E. Our study demonstrates the robustness of digitalMLPA to profile CNAs and to screen point mutations in MM, which could efficiently be used in myeloma diagnostics.
Collapse
Affiliation(s)
- Szabolcs Kosztolányi
- 1st Department of Internal Medicine, Clinical Center, University of Pecs, Pecs, Hungary
| | - Richárd Kiss
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Ambrus Gángó
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | | | - Pál Jáksó
- Department of Pathology, University of Pecs Medical School, Pecs, Hungary
| | - András Matolcsy
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Béla Kajtár
- Department of Pathology, University of Pecs Medical School, Pecs, Hungary
| | - László Pajor
- Department of Pathology, University of Pecs Medical School, Pecs, Hungary
| | - Károly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Csaba Bödör
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Donát Alpár
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary; Department of Pathology, University of Pecs Medical School, Pecs, Hungary.
| |
Collapse
|
316
|
Magierowicz M, Tomowiak C, Leleu X, Poulain S. Working Toward a Genomic Prognostic Classification of Waldenström Macroglobulinemia: C-X-C Chemokine Receptor Type 4 Mutation and Beyond. Hematol Oncol Clin North Am 2018; 32:753-763. [PMID: 30190015 DOI: 10.1016/j.hoc.2018.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Waldenström macroglobulinemia is a rare indolent B-cell lymphoma. Whole-exome sequencing studies have improved our knowledge of the Waldenström macroglobulinemia mutational landscape. The MYD88 L265P mutation is present in nearly 90% of patients with Waldenström macroglobulinemia. CXCR4 mutations are identified in approximately 30% of MYD88L265P cases and have been associated with ibrutinib resistance in clinical trials. Mutations in CD79B, ARID1a, or TP53 were described at lower frequency. Deciphering the earliest initiating lesions and identifying the molecular alterations leading to disease progression currently represent important goals in the future to identify the most relevant targets for precision therapy in Waldenström macroglobulinemia.
Collapse
Affiliation(s)
- Marion Magierowicz
- Laboratory of Hematology, Biology and Pathology Center, CHU of Lille, Lille, France
| | - Cécile Tomowiak
- Department of Hematology, Hospital of the Miletrie, INSERM CIC 1402, CHU of Poitiers, Poitiers, France
| | - Xavier Leleu
- Department of Hematology, Hospital of the Miletrie, INSERM CIC 1402, CHU of Poitiers, Poitiers, France
| | - Stéphanie Poulain
- Laboratory of Hematology, Biology and Pathology Center, CHU of Lille, Lille, France; INSERM UMR S 1172, Team 4, Cancer Research Institute, Lille, France.
| |
Collapse
|
317
|
Subclonal evolution in disease progression from MGUS/SMM to multiple myeloma is characterised by clonal stability. Leukemia 2018; 33:457-468. [PMID: 30046162 PMCID: PMC6365384 DOI: 10.1038/s41375-018-0206-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/01/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022]
Abstract
Multiple myeloma (MM) is a largely incurable haematological malignancy defined by the clonal proliferation of malignant plasma cells (PCs) within the bone marrow. Clonal heterogeneity has recently been established as a feature in MM, however, the subclonal evolution associated with disease progression has not been described. Here, we performed whole-exome sequencing of serial samples from 10 patients, providing new insights into the progression from monoclonal gammopathy of undetermined significance (MGUS) and smouldering MM (SMM), to symptomatic MM. We confirm that intraclonal genetic heterogeneity is a common feature at diagnosis and that the driving events involved in disease progression are more subtle than previously reported. We reveal that MM evolution is mainly characterised by the phenomenon of clonal stability, where the transformed subclonal PC populations identified at MM are already present in the asymptomatic MGUS/SMM stages. Our findings highlight the possibility that PC extrinsic factors may play a role in subclonal evolution and MGUS/SMM to MM progression.
Collapse
|
318
|
Barrio S, Stühmer T, Da-Viá M, Barrio-Garcia C, Lehners N, Besse A, Cuenca I, Garitano-Trojaola A, Fink S, Leich E, Chatterjee M, Driessen C, Martinez-Lopez J, Rosenwald A, Beckmann R, Bargou RC, Braggio E, Stewart AK, Raab MS, Einsele H, Kortüm KM. Spectrum and functional validation of PSMB5 mutations in multiple myeloma. Leukemia 2018; 33:447-456. [PMID: 30026573 DOI: 10.1038/s41375-018-0216-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Despite an increasing number of approved therapies, multiple myeloma (MM) remains an incurable disease and only a small number of patients achieve prolonged disease control. Some genes have been linked with response to commonly used anti-MM compounds, including immunomodulators (IMiDs) and proteasome inhibitors (PIs). In this manuscript, we demonstrate an increased incidence of acquired proteasomal subunit mutations in relapsed MM compared to newly diagnosed disease, underpinning a potential role of point mutations in the clonal evolution of MM. Furthermore, we are first to present and functionally characterize four somatic PSMB5 mutations from primary MM cells identified in a patient under prolonged proteasome inhibition, with three of them affecting the PI-binding pocket S1. We confirm resistance induction through missense mutations not only to Bortezomib, but also, in variable extent, to the next-generation PIs Carfilzomib and Ixazomib. In addition, a negative impact on the proteasome activity is assessed, providing a potential explanation for later therapy-induced eradication of the affected tumor subclones in this patient.
Collapse
Affiliation(s)
- Santiago Barrio
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Thorsten Stühmer
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Matteo Da-Viá
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Nicola Lehners
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrej Besse
- Departement of Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Isabel Cuenca
- Department of Hematology, Hospital Universitario 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Andoni Garitano-Trojaola
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Severin Fink
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Ellen Leich
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Manik Chatterjee
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Driessen
- Departement of Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Joaquin Martinez-Lopez
- Department of Hematology, Hospital Universitario 12 de Octubre, CNIO, Complutense University, Madrid, Spain
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | | | - Ralf C Bargou
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Esteban Braggio
- Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA
| | - A Keith Stewart
- Division of Hematology-Oncology, Mayo Clinic, Scottsdale, AZ, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Marc S Raab
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Hermann Einsele
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - K Martin Kortüm
- Department of Hematology-Oncology, Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
319
|
Towards Molecular Profiling in Multiple Myeloma: A Literature Review and Early Indications of Its Efficacy for Informing Treatment Strategies. Int J Mol Sci 2018; 19:ijms19072087. [PMID: 30021955 PMCID: PMC6073692 DOI: 10.3390/ijms19072087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/14/2018] [Indexed: 12/26/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic malignancy, is characterized by the clonal expansion of plasma cells. Despite dramatic improvements in patients′ survival over the past decade due to advances in therapy exploiting novel molecular targets (immunomodulatory drugs, proteasome inhibitors and monoclonal antibodies), the treatment of relapsed and refractory disease remains challenging. Recent studies confirmed complex, dynamic, and heterogeneous genomic alterations without unifying gene mutations in MM patients. In the current review, we survey recent therapeutic strategies, as well as molecular profiling data on MM, with emphasis on relapsed and refractory cases. A critical appraisal of novel findings and of their potential therapeutic implications will be discussed in detail, along with the author’s own experiences/views.
Collapse
|
320
|
Yang N, Mun YC, Seong CM, Huh HJ, Huh J. Clinicopathological Characteristics of Hyperdiploidy with High-Risk Cytogenetics in Multiple Myeloma. Ann Lab Med 2018; 38:160-164. [PMID: 29214761 PMCID: PMC5736676 DOI: 10.3343/alm.2018.38.2.160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/25/2017] [Accepted: 10/25/2017] [Indexed: 01/06/2023] Open
Abstract
In multiple myeloma (MM), hyperdiploidy (HD) is known to impart longer overall survival. However, it is unclear whether coexistent HD ameliorates the adverse effects of known high-risk cytogenetics in MM patients. To address this issue, we investigated the clinicopathological characteristics of HD with high-risk cytogenetics in MM. Ninety-seven patients with MM were included in the study. For metaphase cytogenetics (MC), unstimulated cells from bone marrow aspirates were cultured for either 24 or 48 hours. To detect HD by interphase fluorescence in situ hybridization (iFISH), we assessed trisomies of chromosomes 5, 7, 9, 11, 15, and 17. Of the 97 MM patients, 40 showed HD. The frequency of co-occurrence of HD and high-risk cytogenetics was 14% (14/97). When the clinicopathological characteristics were compared between the two groups of HD with high-risk cytogenetics vs. non-HD (NHD) with high-risk cytogenetics, the level of beta 2 microglobulin and stage distribution significantly differed (P=0.020, P=0.032, respectively). This study shows that some of the clinicopathological characteristics of MM patients with high-risk cytogenetics differ according to HD or NHD status.
Collapse
Affiliation(s)
- Naery Yang
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Yeung Chul Mun
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Chu Myong Seong
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hee Jin Huh
- Department of Laboratory Medicine, Dongguk University, Ilsan Medical Center, Goyang, Korea.
| | - Jungwon Huh
- Department of Laboratory Medicine, College of Medicine, Ewha Womans University, Seoul, Korea.
| |
Collapse
|
321
|
Saxe D, Seo EJ, Bergeron MB, Han JY. Recent advances in cytogenetic characterization of multiple myeloma. Int J Lab Hematol 2018; 41:5-14. [PMID: 29971938 DOI: 10.1111/ijlh.12882] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022]
Abstract
The detection of cytogenetic abnormalities in multiple myeloma (MM) has received more importance over last years for risk stratification and the new risk-adapted treatment strategies. Conventional G-banding analysis should be included in a routine procedure for the initial diagnostic workup for patients suspected of MM. However, the detection of chromosomal abnormalities in MM by conventional cytogenetics is limited owing to the low proliferative activity of malignant plasma cells as well as the low number of plasma cells in bone marrow specimens. Fluorescence in situ hybridization (FISH) or microarray-based technologies can overcome some of those drawbacks and detect specific target arrangements as well as chromosomal copy number changes. In this review, we will discuss different cytogenetic approaches and compare their strength and weakness to provide genetic information for risk stratification and prediction of outcome in MM patients.
Collapse
Affiliation(s)
- Debra Saxe
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Eul-Ju Seo
- Department of Laboratory Medicine, College of Medicine and Asan Medical Center, University of Ulsan, Seoul, Korea
| | - Melanie Beaulieu Bergeron
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada.,Department of Genetics, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Jin-Yeong Han
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
322
|
Walker BA, Mavrommatis K, Wardell CP, Ashby TC, Bauer M, Davies F, Rosenthal A, Wang H, Qu P, Hoering A, Samur M, Towfic F, Ortiz M, Flynt E, Yu Z, Yang Z, Rozelle D, Obenauer J, Trotter M, Auclair D, Keats J, Bolli N, Fulciniti M, Szalat R, Moreau P, Durie B, Stewart AK, Goldschmidt H, Raab MS, Einsele H, Sonneveld P, San Miguel J, Lonial S, Jackson GH, Anderson KC, Avet-Loiseau H, Munshi N, Thakurta A, Morgan G. A high-risk, Double-Hit, group of newly diagnosed myeloma identified by genomic analysis. Leukemia 2018; 33:159-170. [PMID: 29967379 PMCID: PMC6326953 DOI: 10.1038/s41375-018-0196-8] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022]
Abstract
Patients with newly diagnosed multiple myeloma (NDMM) with high-risk disease are in need of new treatment strategies to improve the outcomes. Multiple clinical, cytogenetic, or gene expression features have been used to identify high-risk patients, each of which has significant weaknesses. Inclusion of molecular features into risk stratification could resolve the current challenges. In a genome-wide analysis of the largest set of molecular and clinical data established to date from NDMM, as part of the Myeloma Genome Project, we have defined DNA drivers of aggressive clinical behavior. Whole-genome and exome data from 1273 NDMM patients identified genetic factors that contribute significantly to progression free survival (PFS) and overall survival (OS) (cumulative R2 = 18.4% and 25.2%, respectively). Integrating DNA drivers and clinical data into a Cox model using 784 patients with ISS, age, PFS, OS, and genomic data, the model has a cumlative R2 of 34.3% for PFS and 46.5% for OS. A high-risk subgroup was defined by recursive partitioning using either a) bi-allelic TP53 inactivation or b) amplification (≥4 copies) of CKS1B (1q21) on the background of International Staging System III, comprising 6.1% of the population (median PFS = 15.4 months; OS = 20.7 months) that was validated in an independent dataset. Double-Hit patients have a dire prognosis despite modern therapies and should be considered for novel therapeutic approaches.
Collapse
Affiliation(s)
- Brian A Walker
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Christopher P Wardell
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - T Cody Ashby
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michael Bauer
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Faith Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Hongwei Wang
- Cancer Research and Biostatistics, Seattle, WA, USA
| | - Pingping Qu
- Cancer Research and Biostatistics, Seattle, WA, USA
| | | | - Mehmet Samur
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Maria Ortiz
- Celgene Institute of Translational Research Europe, Sevilla, Spain
| | | | | | | | | | | | - Matthew Trotter
- Celgene Institute of Translational Research Europe, Sevilla, Spain
| | | | - Jonathan Keats
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | - Raphael Szalat
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Brian Durie
- Cedars-Sinai Samuel Oschin Cancer Center, Los Angeles, CA, USA
| | | | - Hartmut Goldschmidt
- Department of Medicine V, Hematology and Oncology, University Hospital of Heidelberg, Heidelberg, Germany
| | - Marc S Raab
- Department of Medicine V, Hematology and Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Heidelberg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, Wurzburg University, Wurzburg, Germany
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jesus San Miguel
- Clinica Universidad de Navarra, Centro Investigacion Medica Aplicada (CIMA), IDISNA, CIBERONC, Pamplona, Spain
| | - Sagar Lonial
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | | | - Herve Avet-Loiseau
- Centre de Recherche en Cancérologie de Toulouse Institut National de la Santé et de la Recherche Médicale, U1037, Toulouse, France.,L'Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalier Universitaire, Toulouse, France
| | - Nikhil Munshi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Gareth Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
323
|
Ruiz-Heredia Y, Sánchez-Vega B, Onecha E, Barrio S, Alonso R, Martínez-Ávila JC, Cuenca I, Agirre X, Braggio E, Hernández MT, Martínez R, Rosiñol L, Gutierrez N, Martin-Ramos M, Ocio EM, Echeveste MA, de Oteyza JP, Oriol A, Bargay J, Gironella M, Ayala R, Bladé J, Mateos MV, Kortum KM, Stewart K, García-Sanz R, Miguel JS, Lahuerta JJ, Martinez-Lopez J. Mutational screening of newly diagnosed multiple myeloma patients by deep targeted sequencing. Haematologica 2018; 103:e544-e548. [PMID: 29954938 DOI: 10.3324/haematol.2018.188839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Yanira Ruiz-Heredia
- Department of Hematology, Hospital Universitario 12 de Octubre-Hematological Malignancies Clinical Research Unit H120-CNIO, Madrid, Spain
| | - Beatriz Sánchez-Vega
- Department of Hematology, Hospital Universitario 12 de Octubre-Hematological Malignancies Clinical Research Unit H120-CNIO, Madrid, Spain
| | - Esther Onecha
- Department of Hematology, Hospital Universitario 12 de Octubre-Hematological Malignancies Clinical Research Unit H120-CNIO, Madrid, Spain
| | - Santiago Barrio
- Department of Internal Medicine II, University Hospital Würzburg, Germany
| | - Rafael Alonso
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Isabel Cuenca
- Department of Hematology, Hospital Universitario 12 de Octubre-Hematological Malignancies Clinical Research Unit H120-CNIO, Madrid, Spain
| | - Xabier Agirre
- Department of Hematology, Fundación de Investigacion Médica Aplicada (FIMA), Pamplona, Spain
| | - Esteban Braggio
- Department of Hematology, Mayo Clinic, Phoenix/Scottsdale, AZ, USA
| | - Miguel-T Hernández
- Department of Hematology, Hospital Universitario de Canarias, Tenerife, Spain
| | - Rafael Martínez
- Department of Hematology, Hospital Clínico Universitario San Carlos, Madrid, Spain
| | - Laura Rosiñol
- Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Spain
| | - Norma Gutierrez
- Department of Hematology, Hospital Universitario de Salamanca-Centro Investigación del Cáncer and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Marisa Martin-Ramos
- Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Spain
| | - Enrique M Ocio
- Department of Hematology, Hospital Universitario de Salamanca-Centro Investigación del Cáncer and Hospital Universitario-IBSAL, Salamanca, Spain
| | | | | | - Albert Oriol
- Department of Hematology, Hospital Universitario de Salamanca-Centro Investigación del Cáncer and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Joan Bargay
- Department of Hematology, Hospital Sont Llatzer, Palma de Mallorca, Spain
| | - Mercedes Gironella
- Department of Hematology, Hospital Universitario Vall de Hebrón de Barcelona, Spain
| | - Rosa Ayala
- Department of Hematology, Hospital Universitario 12 de Octubre-Hematological Malignancies Clinical Research Unit H120-CNIO, Madrid, Spain
| | - Joan Bladé
- Department of Hematology, Hospital Clinic i Provincial, Institut d'Investigasions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - María-Victoria Mateos
- Department of Hematology, Hospital Universitario de Salamanca-Centro Investigación del Cáncer and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Klaus M Kortum
- Department of Internal Medicine II, University Hospital Würzburg, Germany
| | - Keith Stewart
- Department of Hematology, Mayo Clinic, Phoenix/Scottsdale, AZ, USA
| | - Ramón García-Sanz
- Department of Hematology, Hospital Universitario de Salamanca-Centro Investigación del Cáncer and Hospital Universitario-IBSAL, Salamanca, Spain
| | - Jesús San Miguel
- Department of Hematology, Fundación de Investigacion Médica Aplicada (FIMA), Pamplona, Spain
| | - Juan José Lahuerta
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Joaquín Martinez-Lopez
- Department of Hematology, Hospital Universitario 12 de Octubre-Hematological Malignancies Clinical Research Unit H120-CNIO, Madrid, Spain
| |
Collapse
|
324
|
Morgan GJ, Rasche L. Maintaining therapeutic progress in multiple myeloma by integrating genetic and biological advances into the clinic. Expert Rev Hematol 2018; 11:513-523. [PMID: 29944024 DOI: 10.1080/17474086.2018.1489718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Utilizing advances in genetic and immunologic analysis to segment and direct treatment is potentially a way of maintaining therapeutic progress toward cure in multiple myeloma (MM). This approach works well using clinical segments but can be optimized using recent genetic and immunologic technologies, which have opened the possibility of enhancing risk stratification and disease subclassification. Areas covered: This position paper discusses strategies to segment myeloma into subgroups with distinct risk profiles and distinct targetable lesions are presented. Expert commentary: Risk stratified treatment of MM is already a clinical reality that can be enhanced by the developmental of unified segmentation and testing approaches. Mutation-targeted treatment has proven to be effective against the RAS pathway, but is compromised by intra-clonal and spatiotemporal heterogeneity. Identifying new disease segments based on tumor biology or immunological content of the microenvironment offers an exciting new way to control and even eradicate myeloma clones. Going forward, risk and biologically stratified therapy for myeloma is a promising way of maintaining therapeutic progress, as is precision immunotherapy directed by the cellular context of the bone marrow.
Collapse
Affiliation(s)
- Gareth J Morgan
- a Myeloma Institute , The University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Leo Rasche
- a Myeloma Institute , The University of Arkansas for Medical Sciences , Little Rock , AR , USA
| |
Collapse
|
325
|
Multiple myeloma clonal evolution in homogeneously treated patients. Leukemia 2018; 32:2636-2647. [PMID: 29895955 DOI: 10.1038/s41375-018-0153-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 03/02/2018] [Accepted: 03/09/2018] [Indexed: 02/04/2023]
Abstract
Clonal evolution drives tumor progression, chemoresistance and relapse in cancer. Little is known about clonal selection induced by therapeutic pressure in multiple myeloma. To address this issue, we performed large targeted sequencing of bone marrow plasma cells in 43 multiple myeloma patients at diagnosis and at relapse from exactly the same intensive treatment. The most frequently mutated genes at diagnosis were KRAS (35%), NRAS (28%), DIS3 (16%), BRAF, and LRP1B (12% each). At relapse, the mutational burden was unchanged. Many of the mutations were present at the subclonal level at both time points, including driver ones. According to patients and mutations, we observed different scenarios: selection of a very rare subclone present at diagnosis, appearance, or disappearance of mutations, but also stability. Our data highlight that chemoresistance and relapse could be induced by newly acquired mutations in myeloma drivers but also by (sub)clonal mutations preexisting to the treatment. Importantly, no specific mutation or rearrangement was observed at relapse, demonstrating that intensive treatment has a nonspecific effect on clonal selection in multiple myeloma. Finally, we identified 22 cases of biallelic event, including a double event deletion 17p/TP53mut.
Collapse
|
326
|
Soekojo CY, de Mel S, Ooi M, Yan B, Chng WJ. Potential Clinical Application of Genomics in Multiple Myeloma. Int J Mol Sci 2018; 19:ijms19061721. [PMID: 29890777 PMCID: PMC6032230 DOI: 10.3390/ijms19061721] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/02/2018] [Accepted: 06/07/2018] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma is a heterogeneous disease with different characteristics, and genetic aberrations play important roles in this heterogeneity. Studies have shown that these genetic aberrations are crucial in prognostication and response assessment; recent efforts have focused on their possible therapeutic implications. Despite many emerging studies being published, the best way to incorporate these results into clinical practice remains unclear. In this review paper we describe the different genomic techniques available, including the latest advancements, and discuss the potential clinical application of genomics in multiple myeloma.
Collapse
Affiliation(s)
- Cinnie Yentia Soekojo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Sanjay de Mel
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Melissa Ooi
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
| | - Benedict Yan
- Department of Laboratory Medicine, National University Hospital, National University Health System, 5 Lower Kent Ridge Road, Singapore 119074, Singapore.
| | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, 1E Kent Ridge Road, Singapore 119228, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore,14 Medical Drive, Singapore 117599, Singapore.
| |
Collapse
|
327
|
Identification of novel mutational drivers reveals oncogene dependencies in multiple myeloma. Blood 2018; 132:587-597. [PMID: 29884741 DOI: 10.1182/blood-2018-03-840132] [Citation(s) in RCA: 337] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/04/2018] [Indexed: 12/11/2022] Open
Abstract
Understanding the profile of oncogene and tumor suppressor gene mutations with their interactions and impact on the prognosis of multiple myeloma (MM) can improve the definition of disease subsets and identify pathways important in disease pathobiology. Using integrated genomics of 1273 newly diagnosed patients with MM, we identified 63 driver genes, some of which are novel, including IDH1, IDH2, HUWE1, KLHL6, and PTPN11 Oncogene mutations are significantly more clonal than tumor suppressor mutations, indicating they may exert a bigger selective pressure. Patients with more driver gene abnormalities are associated with worse outcomes, as are identified mechanisms of genomic instability. Oncogenic dependencies were identified between mutations in driver genes, common regions of copy number change, and primary translocation and hyperdiploidy events. These dependencies included associations with t(4;14) and mutations in FGFR3, DIS3, and PRKD2; t(11;14) with mutations in CCND1 and IRF4; t(14;16) with mutations in MAF, BRAF, DIS3, and ATM; and hyperdiploidy with gain 11q, mutations in FAM46C, and MYC rearrangements. These associations indicate that the genomic landscape of myeloma is predetermined by the primary events upon which further dependencies are built, giving rise to a nonrandom accumulation of genetic hits. Understanding these dependencies may elucidate potential evolutionary patterns and lead to better treatment regimens.
Collapse
|
328
|
Mansilla C, Soria E, Vallejo M, Valiente A, Perez-Juana A, Zabalza A, Hurtado G, Sala F, Ramírez N. Combined Selection System to Lower the Cutoff for Plasma Cell Enrichment Applied to iFISH Analysis in Multiple Myeloma. Transl Oncol 2018; 11:647-652. [PMID: 29604510 PMCID: PMC6054590 DOI: 10.1016/j.tranon.2018.02.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma (MM) is a very heterogeneous disease, characterized by multiple cytogenetic aberrations on plasma cells (PC) that have been traditionally used to predict the outcome of the disease. A mayor issue on the analysis of PC is the sometimes low infiltration of these cells in the bone marrow that hampers cytogenetic studies. To solve this problem we have optimized a selection strategy based on PC immunomagnetic isolation that has allowed us to lower to 1% the minimal PC infiltration requirement without loss of purity, enabling to perform genetic analysis. In this study, we have analyzed 153 bone marrow samples of patients suspected of MM, collected from February 2015 to May 2017 by the Genetics service of the Complejo Hospitalario de Navarra. Clinical characteristics of the patients and PC immunophenotyping, conventional cytogenetics and interphase fluorescence in situ hybridization (iFISH) analyses have been assessed on these samples. In our cohort 90% of the samples had cytogenetic abnormalities, among them 50% presented immunoglobulin rearrangements, 41.9% showed 1q gains, 29.7% showed 1p deletions and 33% presented TP53 deletion.
Collapse
Affiliation(s)
- Cristina Mansilla
- Oncohematology Research Group, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, IdiSNA
| | - Elena Soria
- Oncohematology Research Group, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, IdiSNA
| | - Miren Vallejo
- Oncohematology Research Group, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, IdiSNA
| | - Alberto Valiente
- Genetics Service, Complejo Hospitalario de Navarra, IdiSNA (Navarra's Health Research Institute), Pamplona, Spain
| | - Aránzazu Perez-Juana
- Genetics Service, Complejo Hospitalario de Navarra, IdiSNA (Navarra's Health Research Institute), Pamplona, Spain
| | - Amaya Zabalza
- Oncohematology Research Group, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, IdiSNA; Hematology Department, Complejo Hospitalario de Navarra, IdiSNA, Pamplona, Spain
| | - Guillermina Hurtado
- Cytometry Service, Hematology Department, Complejo Hospitalario de Navarra, IdiSNA, Pamplona, Spain
| | - Francisco Sala
- Cytometry Service, Hematology Department, Complejo Hospitalario de Navarra, IdiSNA, Pamplona, Spain
| | - Natalia Ramírez
- Oncohematology Research Group, Navarrabiomed, Complejo Hospitalario de Navarra, Universidad Pública de Navarra, IdiSNA.
| |
Collapse
|
329
|
Derenzini E, Rossi A, Treré D. Treating hematological malignancies with drugs inhibiting ribosome biogenesis: when and why. J Hematol Oncol 2018; 11:75. [PMID: 29855342 PMCID: PMC5984324 DOI: 10.1186/s13045-018-0609-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/26/2018] [Indexed: 01/05/2023] Open
Abstract
It is well known that chemotherapy can cure only some cancers in advanced stage, mostly those with an intact p53 pathway. Hematological cancers such as lymphoma and certain forms of leukemia are paradigmatic examples of such scenario. Recent evidence indicates that the efficacy of many of the alkylating and intercalating agents, antimetabolites, topoisomerase, and kinase inhibitors used in cancer therapy is largely due to p53 stabilization and activation consequent to the inhibition of ribosome biogenesis. In this context, innovative drugs specifically hindering ribosome biogenesis showed preclinical activity and are currently in early clinical development in hematological malignancies. The mechanism of p53 stabilization after ribosome biogenesis inhibition is a multistep process, depending on specific factors that can be altered in tumor cells, which can affect the antitumor efficacy of ribosome biogenesis inhibitors (RiBi). In the present review, the basic mechanisms underlying the anticancer activity of RiBi are discussed based on the evidence deriving from available preclinical and clinical studies, with the purpose of defining when and why the treatment with drugs inhibiting ribosomal biogenesis could be highly effective in hematological malignancies.
Collapse
Affiliation(s)
- Enrico Derenzini
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy.
| | - Alessandra Rossi
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy
| | - Davide Treré
- DIMES, Università di Bologna, Via Massarenti 9, Bologna, Italy.
| |
Collapse
|
330
|
Bolli N, Biancon G, Moarii M, Gimondi S, Li Y, de Philippis C, Maura F, Sathiaseelan V, Tai YT, Mudie L, O'Meara S, Raine K, Teague JW, Butler AP, Carniti C, Gerstung M, Bagratuni T, Kastritis E, Dimopoulos M, Corradini P, Anderson KC, Moreau P, Minvielle S, Campbell PJ, Papaemmanuil E, Avet-Loiseau H, Munshi NC. Analysis of the genomic landscape of multiple myeloma highlights novel prognostic markers and disease subgroups. Leukemia 2018; 32:2604-2616. [PMID: 29789651 PMCID: PMC6092251 DOI: 10.1038/s41375-018-0037-9] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/28/2017] [Accepted: 11/10/2017] [Indexed: 12/19/2022]
Abstract
In multiple myeloma, next-generation sequencing (NGS) has expanded our knowledge of genomic lesions, and highlighted a dynamic and heterogeneous composition of the tumor. Here we used NGS to characterize the genomic landscape of 418 multiple myeloma cases at diagnosis and correlate this with prognosis and classification. Translocations and copy number abnormalities (CNAs) had a preponderant contribution over gene mutations in defining the genotype and prognosis of each case. Known and novel independent prognostic markers were identified in our cohort of proteasome inhibitor and immunomodulatory drug-treated patients with long follow-up, including events with context-specific prognostic value, such as deletions of the PRDM1 gene. Taking advantage of the comprehensive genomic annotation of each case, we used innovative statistical approaches to identify potential novel myeloma subgroups. We observed clusters of patients stratified based on the overall number of mutations and number/type of CNAs, with distinct effects on survival, suggesting that extended genotype of multiple myeloma at diagnosis may lead to improved disease classification and prognostication.
Collapse
Affiliation(s)
- Niccolo Bolli
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy.,Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Giulia Biancon
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Matahi Moarii
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Silvia Gimondi
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | - Yilong Li
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Chiara de Philippis
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy.,Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Maura
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy
| | | | - Yu-Tzu Tai
- Harvard Medical School, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laura Mudie
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Sarah O'Meara
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Keiran Raine
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Jon W Teague
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Adam P Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Cristiana Carniti
- Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Moritz Gerstung
- European Bioinformatics Institute, Computational and Cancer Biology, Cambridge, UK
| | - Tina Bagratuni
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios Dimopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, Athens, Greece
| | - Paolo Corradini
- Department of Oncology and Onco-Hematology, University of Milan, Milan, Italy.,Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Kenneth C Anderson
- Harvard Medical School, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philippe Moreau
- Department of Hematology, University Hospital Hôtel-Dieu, Nantes, France
| | - Stephane Minvielle
- Department of Hematology, University Hospital Hôtel-Dieu, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Peter J Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Herve Avet-Loiseau
- Institute Universitaire du Cancer de Toulouse Oncopole, Toulouse, France.,University Hospital, Toulouse, France
| | - Nikhil C Munshi
- Harvard Medical School, LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
331
|
Roy P, Sarkar UA, Basak S. The NF-κB Activating Pathways in Multiple Myeloma. Biomedicines 2018; 6:biomedicines6020059. [PMID: 29772694 PMCID: PMC6027071 DOI: 10.3390/biomedicines6020059] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple myeloma(MM), an incurable plasma cell cancer, represents the second most prevalent hematological malignancy. Deregulated activity of the nuclear factor kappaB (NF-κB) family of transcription factors has been implicated in the pathogenesis of multiple myeloma. Tumor microenvironment-derived cytokines and cancer-associated genetic mutations signal through the canonical as well as the non-canonical arms to activate the NF-κB system in myeloma cells. In fact, frequent engagement of both the NF-κB pathways constitutes a distinguishing characteristic of myeloma. In turn, NF-κB signaling promotes proliferation, survival and drug-resistance of myeloma cells. In this review article, we catalog NF-κB activating genetic mutations and microenvironmental cues associated with multiple myeloma. We then describe how the individual canonical and non-canonical pathways transduce signals and contribute towards NF-κB -driven gene-expressions in healthy and malignant cells. Furthermore, we discuss signaling crosstalk between concomitantly triggered NF-κB pathways, and its plausible implication for anomalous NF-κB activation and NF-κB driven pro-survival gene-expressions in multiple myeloma. Finally, we propose that mechanistic understanding of NF-κB deregulations may provide for improved therapeutic and prognostic tools in multiple myeloma.
Collapse
Affiliation(s)
- Payel Roy
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Uday Aditya Sarkar
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| | - Soumen Basak
- Systems Immunology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India.
| |
Collapse
|
332
|
Ryland GL, Jones K, Chin M, Markham J, Aydogan E, Kankanige Y, Caruso M, Guinto J, Dickinson M, Prince HM, Yong K, Blombery P. Novel genomic findings in multiple myeloma identified through routine diagnostic sequencing. J Clin Pathol 2018; 71:895-899. [PMID: 29760015 DOI: 10.1136/jclinpath-2018-205195] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/20/2018] [Accepted: 04/20/2018] [Indexed: 01/10/2023]
Abstract
AIMS Multiple myeloma is a genomically complex haematological malignancy with many genomic alterations recognised as important in diagnosis, prognosis and therapeutic decision making. Here, we provide a summary of genomic findings identified through routine diagnostic next-generation sequencing at our centre. METHODS A cohort of 86 patients with multiple myeloma underwent diagnostic sequencing using a custom hybridisation-based panel targeting 104 genes. Sequence variants, genome-wide copy number changes and structural rearrangements were detected using an inhouse-developed bioinformatics pipeline. RESULTS At least one mutation was found in 69 (80%) patients. Frequently mutated genes included TP53 (36%), KRAS (22.1%), NRAS (15.1%), FAM46C/DIS3 (8.1%) and TET2/FGFR3 (5.8%), including multiple mutations not previously described in myeloma. Importantly we observed TP53 mutations in the absence of a 17 p deletion in 8% of the cohort, highlighting the need for sequencing-based assessment in addition to cytogenetics to identify these high-risk patients. Multiple novel copy number changes and immunoglobulin heavy chain translocations are also discussed. CONCLUSIONS Our results demonstrate that many clinically relevant genomic findings remain in multiple myeloma which have not yet been identified through large-scale sequencing efforts, and provide important mechanistic insights into plasma cell pathobiology.
Collapse
Affiliation(s)
- Georgina L Ryland
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kate Jones
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Melody Chin
- Department of Haematology, University College London Cancer Institute, London, UK
| | - John Markham
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Elle Aydogan
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Yamuna Kankanige
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Marisa Caruso
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jerick Guinto
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael Dickinson
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - H Miles Prince
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kwee Yong
- Department of Haematology, University College London Cancer Institute, London, UK
| | - Piers Blombery
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
333
|
Zhang X, Li B, Han H, Song S, Xu H, Hong Y, Yi N, Zhuang W. Predicting multi-level drug response with gene expression profile in multiple myeloma using hierarchical ordinal regression. BMC Cancer 2018; 18:551. [PMID: 29747599 PMCID: PMC5946496 DOI: 10.1186/s12885-018-4483-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/07/2018] [Indexed: 11/22/2022] Open
Abstract
Background Multiple myeloma (MM), like other cancers, is caused by the accumulation of genetic abnormalities. Heterogeneity exists in the patients’ response to treatments, for example, bortezomib. This urges efforts to identify biomarkers from numerous molecular features and build predictive models for identifying patients that can benefit from a certain treatment scheme. However, previous studies treated the multi-level ordinal drug response as a binary response where only responsive and non-responsive groups are considered. Methods It is desirable to directly analyze the multi-level drug response, rather than combining the response to two groups. In this study, we present a novel method to identify significantly associated biomarkers and then develop ordinal genomic classifier using the hierarchical ordinal logistic model. The proposed hierarchical ordinal logistic model employs the heavy-tailed Cauchy prior on the coefficients and is fitted by an efficient quasi-Newton algorithm. Results We apply our hierarchical ordinal regression approach to analyze two publicly available datasets for MM with five-level drug response and numerous gene expression measures. Our results show that our method is able to identify genes associated with the multi-level drug response and to generate powerful predictive models for predicting the multi-level response. Conclusions The proposed method allows us to jointly fit numerous correlated predictors and thus build efficient models for predicting the multi-level drug response. The predictive model for the multi-level drug response can be more informative than the previous approaches. Thus, the proposed approach provides a powerful tool for predicting multi-level drug response and has important impact on cancer studies. Electronic supplementary material The online version of this article (10.1186/s12885-018-4483-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinyan Zhang
- Department of Biostatistics, Jiann-Ping Hsu College of Public Health, Georgia Southern University, Statesboro, GA, USA
| | - Bingzong Li
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Huiying Han
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Sha Song
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Hongxia Xu
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Yating Hong
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Wenzhuo Zhuang
- Department of Cell Biology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
334
|
Kumar SK, Rajkumar SV. The multiple myelomas — current concepts in cytogenetic classification and therapy. Nat Rev Clin Oncol 2018; 15:409-421. [DOI: 10.1038/s41571-018-0018-y] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
335
|
Hoang PH, Dobbins SE, Cornish AJ, Chubb D, Law PJ, Kaiser M, Houlston RS. Whole-genome sequencing of multiple myeloma reveals oncogenic pathways are targeted somatically through multiple mechanisms. Leukemia 2018; 32:2459-2470. [PMID: 29654271 PMCID: PMC6224406 DOI: 10.1038/s41375-018-0103-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/16/2018] [Accepted: 03/05/2018] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a biologically heterogeneous malignancy, however, the mechanisms underlying this complexity are incompletely understood. We report an analysis of the whole-genome sequencing of 765 MM patients from CoMMpass. By employing promoter capture Hi-C in naïve B-cells, we identify cis-regulatory elements (CREs) that represent a highly enriched subset of the non-coding genome in which to search for driver mutations. We identify regulatory regions whose mutation significantly alters the expression of genes as candidate non-coding drivers, including copy number variation (CNV) at CREs of MYC and single-nucleotide variants (SNVs) in a PAX5 enhancer. To better inform the interplay between non-coding driver mutations with other driver mechanisms, and their respective roles in oncogenic pathways, we extended our analysis identifying coding drivers in 40 genes, including 11 novel candidates. We demonstrate the same pathways can be targeted by coding and non-coding mutations; exemplified by IRF4 and PRDM1, along with BCL6 and PAX5, genes that are central to plasma cell differentiation. This study reveals new insights into the complex genetic alterations driving MM development and an enhanced understanding of oncogenic pathways.
Collapse
Affiliation(s)
- Phuc H Hoang
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK.,Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Sara E Dobbins
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Alex J Cornish
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Daniel Chubb
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Martin Kaiser
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK. .,Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
| |
Collapse
|
336
|
Genomic discovery and clonal tracking in multiple myeloma by cell-free DNA sequencing. Leukemia 2018; 32:1838-1841. [PMID: 29749395 DOI: 10.1038/s41375-018-0115-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/05/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
|
337
|
Search for rare protein altering variants influencing susceptibility to multiple myeloma. Oncotarget 2018; 8:36203-36210. [PMID: 28404951 PMCID: PMC5482649 DOI: 10.18632/oncotarget.15874] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/28/2017] [Indexed: 12/12/2022] Open
Abstract
The genetic basis underlying the inherited risk of developing multiple myeloma (MM) is largely unknown. To examine the impact of rare protein altering variants on the risk of developing MM we analyzed high-coverage exome sequencing data on 513 MM cases and 1,569 healthy controls, performing both single variant and gene burden tests. We did not identify any recurrent coding low-frequency alleles (1–5%) with moderate effect that were statistically associated with MM. In a gene burden analysis we did however identify a promising relationship between variation in the marrow kinetochore microtubule stromal gene KIF18A, which plays a role in control mitotic chromosome positioning dynamics, and risk of MM (P =3.6×10−6). Further analysis showed KIF18A displays a distinct pattern of expression across molecular subgroups of MM as well as being associated with patient survival. Our results inform future study design and provide a resource for contextualizing the impact of candidate MM susceptibility genes.
Collapse
|
338
|
Kinase domain activation through gene rearrangement in multiple myeloma. Leukemia 2018; 32:2435-2444. [PMID: 29654269 PMCID: PMC6224403 DOI: 10.1038/s41375-018-0108-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/08/2018] [Accepted: 03/02/2018] [Indexed: 12/28/2022]
Abstract
Chromosomal rearrangements that result in oncogenic kinase activation are present in many solid and hematological malignancies, but none have been reported in multiple myeloma (MM). Here we analyzed 1421 samples from 958 myeloma patients using a targeted assay and detected fusion genes in 1.5% of patients. These fusion genes were in-frame and the majority of them contained kinase domains from either receptor tyrosine kinases (ALK, ROS1, NTRK3, and FGFR1) or cytoplasmic kinases (BRAF, MAP3K14, and MAPK14), which would result in the activation of MEK/ERK, NF-κB, or inflammatory signaling pathways. Fusion genes were present in smoldering MM, newly diagnosed MM, and relapse patient samples indicating they are not solely late events. Most fusion genes were subclonal in nature, but one EML4-ALK fusion was clonal indicating it is a driver of disease pathogenesis. Samples with fusions of receptor tyrosine kinases were not found in conjunction with clonal Ras/Raf mutations indicating a parallel mechanism of MEK/ERK pathway activation. Fusion genes involving MAP3K14 (NIK), which regulates the NF-κB pathway, were detected as were t(14;17) rearrangements involving NIK in 2% of MM samples. Activation of kinases in myeloma through rearrangements presents an opportunity to use treatments existing in other cancers.
Collapse
|
339
|
White BS, Lanc I, O'Neal J, Gupta H, Fulton RS, Schmidt H, Fronick C, Belter EA, Fiala M, King J, Ahmann GJ, DeRome M, Mardis ER, Vij R, DiPersio JF, Levy J, Auclair D, Tomasson MH. A multiple myeloma-specific capture sequencing platform discovers novel translocations and frequent, risk-associated point mutations in IGLL5. Blood Cancer J 2018; 8:35. [PMID: 29563506 PMCID: PMC5862875 DOI: 10.1038/s41408-018-0062-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/10/2017] [Accepted: 12/18/2017] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma (MM) is a disease of copy number variants (CNVs), chromosomal translocations, and single-nucleotide variants (SNVs). To enable integrative studies across these diverse mutation types, we developed a capture-based sequencing platform to detect their occurrence in 465 genes altered in MM and used it to sequence 95 primary tumor-normal pairs to a mean depth of 104×. We detected cases of hyperdiploidy (23%), deletions of 1p (8%), 6q (21%), 8p (17%), 14q (16%), 16q (22%), and 17p (4%), and amplification of 1q (19%). We also detected IGH and MYC translocations near expected frequencies and non-silent SNVs in NRAS (24%), KRAS (21%), FAM46C (17%), TP53 (9%), DIS3 (9%), and BRAF (3%). We discovered frequent mutations in IGLL5 (18%) that were mutually exclusive of RAS mutations and associated with increased risk of disease progression (p = 0.03), suggesting that IGLL5 may be a stratifying biomarker. We identified novel IGLL5/IGH translocations in two samples. We subjected 15 of the pairs to ultra-deep sequencing (1259×) and found that although depth correlated with number of mutations detected (p = 0.001), depth past ~300× added little. The platform provides cost-effective genomic analysis for research and may be useful in individualizing treatment decisions in clinical settings.
Collapse
Affiliation(s)
- Brian S White
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA.,Sage Bionetworks, Seattle, WA, 91809, USA
| | - Irena Lanc
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Julie O'Neal
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Harshath Gupta
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, 63108, MO, USA
| | - Heather Schmidt
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, 63108, MO, USA
| | - Catrina Fronick
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, 63108, MO, USA
| | - Edward A Belter
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, 63108, MO, USA
| | - Mark Fiala
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Justin King
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Greg J Ahmann
- Division of Hematology-Oncology, Mayo Clinic, Rochester, 55905, MN, USA
| | - Mary DeRome
- Multiple Myeloma Research Foundation, Norwalk, CT, 06851, USA
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, 63108, MO, USA.,Genomics Institute, Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Ravi Vij
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - John F DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA
| | - Joan Levy
- Multiple Myeloma Research Foundation, Norwalk, CT, 06851, USA.,Chordoma Foundation, Durham, NC, 27702, USA
| | - Daniel Auclair
- Multiple Myeloma Research Foundation, Norwalk, CT, 06851, USA
| | - Michael H Tomasson
- Department of Medicine, Washington University School of Medicine, St. Louis, 63110, MO, USA. .,Division of Hematology, Oncology and Bone Marrow Transplantation, 5204 MERF, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
340
|
Evaluation of in vitro effects of various targeted drugs on plasma cells and putative neoplastic stem cells in patients with multiple myeloma. Oncotarget 2018; 7:65627-65642. [PMID: 27582537 PMCID: PMC5323180 DOI: 10.18632/oncotarget.11593] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 08/13/2016] [Indexed: 01/08/2023] Open
Abstract
Multiple myeloma (MM) is a malignancy characterized by monoclonal paraproteinemia and tissue plasmocytosis. In advanced MM cytopenia and osteopathy may occur. Although several effective treatment strategies have been developed in recent years, there is still a need to identify new drug targets and to develop more effective therapies for patients with advanced MM. We examined the effects of 15 targeted drugs on growth and survival of primary MM cells and 5 MM cell lines (MM.1S, NCI-H929, OPM-2, RPMI-8226, U-266). The PI3-kinase blocker BEZ235, the pan-BCL-2 inhibitor obatoclax, the Hsp90-targeting drug 17AAG, and the Polo-like kinase-1 inhibitor BI2536, were found to exert major growth-inhibitory effects in all 5 MM cell lines tested. Moreover, these drugs suppressed the in vitro proliferation of primary bone marrow-derived MM cells and induced apoptosis at pharmacologic drug concentrations. Apoptosis-inducing effects were not only seen in the bulk of MM cells but also in MM stem cell-containing CD138−/CD20+/CD27+ memory B-cell fractions. Synergistic growth-inhibitory effects were observed in MM cell lines using various drug combinations, including 17AAG+BI2536 in MM.1S, OPM-2, RPMI-8226, and U-266 cells, 17AAG+BEZ235 in MM.1S, OPM-2, RPMI-8226, and U-266 cells, 17AAG+obatoclax in MM.1S, NCI-H929, OPM-2, and RPMI-8226 cells, BI2536+BEZ235 in MM.1S, NCI-H929, OPM-2, and RPMI-8226 cells, BI2536+obatoclax in MM.1S, OPM-2 and RPMI-8226 cells, and BEZ235+obatoclax in MM.1S and RPMI-8226 cells. Together, our data show that various targeted drugs induce profound and often synergistic anti-neoplastic effects in MM cells which may have clinical implications and may contribute to the development of novel treatment strategies in advanced MM.
Collapse
|
341
|
Landgren O, Lu SX, Hultcrantz M. MRD Testing in Multiple Myeloma: The Main Future Driver for Modern Tailored Treatment. Semin Hematol 2018; 55:44-50. [PMID: 29759154 DOI: 10.1053/j.seminhematol.2018.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/02/2018] [Indexed: 11/11/2022]
Abstract
The past decade, several highly efficacious drugs have been approved for the treatment of multiple myeloma. Many of these newer drugs are less toxic than older chemotherapy drugs. Using modern combination therapy in newly diagnosed multiple myeloma patients, high proportions of newly diagnosed multiple myeloma patients obtain minimal residual disease (MRD) negativity and MRD testing has rapidly become an integral part of clinical trials focusing on patients in this setting. Only recently, MRD negativity was reported in clinical trials focusing on older newly diagnosed multiple myeloma patients (ie, nontransplant candidates), as well as studies focusing on patients with relapsed or refractory multiple myeloma. In the past, deeper responses were rarely seen in these patient categories due to inferior therapies and lack of MRD assays. The reason for the rapidly increased interest in MRD testing in all types of clinical trials is the fact that MRD negativity is closely correlated with longer progression-free survival which has been documented in recent meta-analyses. Consequently, MRD negativity has the potential to soon become a regulatory surrogate end-point for drug approval. This review dissects and discusses current data on MRD in multiple myeloma, it outlines new hypotheses, which can be tested in future clinical studies, and it discusses opportunities and future avenues for translational research. The goal of this article is to stimulate critical analysis of our current treatment landscape and development of future translational research involving MRD testing.
Collapse
Affiliation(s)
- Ola Landgren
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY.
| | - Sydney X Lu
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Malin Hultcrantz
- Department of Medicine, Myeloma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
342
|
Accardi F, Marchica V, Mancini C, Maredi E, Racano C, Notarfranchi L, Martorana D, Storti P, Martella E, Palma BD, Craviotto L, Filippo MD, Percesepe A, Aversa F, Giuliani N. Neurofibromatosis type I and multiple myeloma coexistence: A possible link? Hematol Rep 2018; 10:7457. [PMID: 29721253 PMCID: PMC5907645 DOI: 10.4081/hr.2018.7457] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/28/2018] [Indexed: 11/22/2022] Open
Abstract
The association between Neurofibromatosis type I (NF1) and multiple myeloma (MM), a plasma cell, dyscrasia is very rare. Here we put to the attention of the scientific community two new cases. The first one is a patient with active MM whereas the second with smoldering MM. Both patients present typical features of NF1 but skeletal alterations were present only in the second case including dysplasia, marked scoliosis and osteoporosis. MM osteolytic lesions were absent in both patients. In addition to the clinical diagnosis of NF1, a molecular testing for NF1 gene mutations has been performed finding that patient one was heterozygous for the c.6855C>A (Tyr2285Ter) mutation, while patient two was heterozygous for the c.7838dupC (Lys2614GlufsTer20) mutation. The two mutations were diagnosed both in genomic DNA from peripheral blood and from MM cells. The potential link between NF1 mutation and the increased risk of MM is discussed in the report.
Collapse
Affiliation(s)
- Fabrizio Accardi
- Department of Medicine and Surgery, University of Parma.,Hematology Unit, Parma University Hospital
| | | | | | - Elena Maredi
- Pediatric Orthopedics, Rizzoli Orthopedic Institute, Bologna
| | | | - Laura Notarfranchi
- Department of Medicine and Surgery, University of Parma.,Hematology Unit, Parma University Hospital
| | | | - Paola Storti
- Department of Medicine and Surgery, University of Parma
| | | | - Benedetta Dalla Palma
- Department of Medicine and Surgery, University of Parma.,Hematology Unit, Parma University Hospital
| | - Luisa Craviotto
- Department of Medicine and Surgery, University of Parma.,Hematology Unit, Parma University Hospital
| | | | | | - Franco Aversa
- Department of Medicine and Surgery, University of Parma.,Hematology Unit, Parma University Hospital
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma.,Hematology Unit, Parma University Hospital
| |
Collapse
|
343
|
DNMTi/HDACi combined epigenetic targeted treatment induces reprogramming of myeloma cells in the direction of normal plasma cells. Br J Cancer 2018; 118:1062-1073. [PMID: 29500406 PMCID: PMC5931098 DOI: 10.1038/s41416-018-0025-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/10/2018] [Accepted: 01/15/2018] [Indexed: 01/18/2023] Open
Abstract
Background Multiple myeloma (MM) is the second most common hematologic malignancy. Aberrant epigenetic modifications have been reported in MM and could be promising therapeutic targets. As response rates are overall limited but deep responses occur, it is important to identify those patients who could indeed benefit from epigenetic-targeted therapy. Methods Since HDACi and DNMTi combination have potential therapeutic value in MM, we aimed to build a GEP-based score that could be useful to design future epigenetic-targeted combination trials. In addition, we investigated the changes in GEP upon HDACi/DNMTi treatment. Results We report a new gene expression-based score to predict MM cell sensitivity to the combination of DNMTi/HDACi. A high Combo score in MM patients identified a group with a worse overall survival but a higher sensitivity of their MM cells to DNMTi/HDACi therapy compared to a low Combo score. In addition, treatment with DNMTi/HDACi downregulated IRF4 and MYC expression and appeared to induce a mature BMPC plasma cell gene expression profile in myeloma cell lines. Conclusion In conclusion, we developed a score for the prediction of primary MM cell sensitivity to DNMTi/HDACi and found that this combination could be beneficial in high-risk patients by targeting proliferation and inducing maturation.
Collapse
|
344
|
Kreitman RJ, Arons E. Update on hairy cell leukemia. CLINICAL ADVANCES IN HEMATOLOGY & ONCOLOGY : H&O 2018; 16:205-215. [PMID: 29742076 PMCID: PMC6290912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Hairy cell leukemia (HCL) is a chronic B-cell malignancy with multiple treatment options, including several that are investigational. Patients present with pancytopenia and splenomegaly, owing to the infiltration of leukemic cells expressing CD22, CD25, CD20, CD103, tartrate-resistant acid phosphatase (TRAP), annexin A1 (ANXA1), and the BRAF V600E mutation. A variant lacking CD25, ANXA1, TRAP, and the BRAF V600E mutation, called HCLv, is more aggressive and is classified as a separate disease. A molecularly defined variant expressing unmutated immunoglobulin heavy variable 4-34 (IGHV4-34) is also aggressive, lacks the BRAF V600E mutation, and has a phenotype of HCL or HCLv. The standard first-line treatment, which has remained unchanged for the past 25 to 30 years, is single-agent therapy with a purine analogue, either cladribine or pentostatin. This approach produces a high rate of complete remission. Residual traces of HCL cells, referred to as minimal residual disease, are present in most patients and cause frequent relapse. Repeated treatment with a purine analogue can restore remission, but at decreasing rates and with increasing cumulative toxicity. Rituximab has limited activity as a single agent but achieves high complete remission rates without minimal residual disease when combined with purine analogues, albeit with chemotherapy-associated toxicity. Investigational nonchemotherapy options include moxetumomab pasudotox, which targets CD22; vemurafenib or dabrafenib, each of which targets the BRAF V600E protein; trametinib, which targets mitogen-activated protein kinase enzyme (MEK); and ibrutinib, which targets Bruton tyrosine kinase (BTK).
Collapse
Affiliation(s)
- Robert J Kreitman
- National Cancer Institute's Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Evgeny Arons
- National Cancer Institute's Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
345
|
Waldschmidt JM, Anand P, Knoechel B, Lohr JG. Comprehensive characterization of circulating and bone marrow-derived multiple myeloma cells at minimal residual disease. Semin Hematol 2018; 55:33-37. [PMID: 29759150 DOI: 10.1053/j.seminhematol.2018.02.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 01/04/2023]
Abstract
The presence or absence of minimal residual disease (MRD) in patients with multiple myeloma (MM) has emerged as a useful marker to determine the depth of remission. MRD negativity as an endpoint has been shown to be associated with improved progression-free survival in many studies. MRD detection is therefore part of numerous clinical trial protocols for MM. At the present time, two methodologies are most widely accepted for MRD detection: (1) multicolor flow cytometry and (2) next-generation sequencing-based clonotype detection. While both of those methodologies enable accurate quantification of MRD in the bone marrow (BM), with sensitivity as low as 10-5 to 10-6, there are several limitations to these methods. First, these approaches reveal the presence or absence of MRD but provide limited molecular information about MM. More comprehensive characterization of MM cells at the MRD stage may identify molecular mechanisms of drug resistance. Second, MRD detection in the BM is typically performed at one time point only, but more frequent detection may define the duration of the MRD status and thus refine its prognostic value. Third, less-invasive approaches that avoid the discomfort and risk associated with BM biopsy would be highly desirable, especially in elderly or frail patients. "Liquid biopsy" for the detection and characterization of circulating MM cells may address these issues. Although MRD detection in the peripheral blood at the same sensitivity as in the BM may be challenging, the identification of patients who do not achieve MRD negativity might reduce the need for BM biopsies. Here, we give an overview of approaches that have been described to detect and characterize MM cells when they occur at very low frequencies in the peripheral blood or in the BM, emphasizing recently described next-generation sequencing approaches for more comprehensive characterization of circulating MM cells.
Collapse
Affiliation(s)
- Johannes M Waldschmidt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Praveen Anand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Birgit Knoechel
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Jens G Lohr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
| |
Collapse
|
346
|
Jovanović KK, Roche-Lestienne C, Ghobrial IM, Facon T, Quesnel B, Manier S. Targeting MYC in multiple myeloma. Leukemia 2018; 32:1295-1306. [PMID: 29467490 DOI: 10.1038/s41375-018-0036-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 11/11/2017] [Accepted: 11/16/2017] [Indexed: 12/18/2022]
Abstract
Multiple myeloma (MM) is a plasma cell tumor marked by clonal evolution and preceded by a premalignant stage, which progresses via molecular pathway deregulation, including MYC activation. This activation relates to translocation or gain of the MYC locus and deregulation of upstream pathways such as IRF4, DIS3/LIN28B/let-7, or MAPK. Precision medicine is an approach to predict more accurately which treatment strategies for a particular disease will work in which groups of patients, in contrast to a "one-size-fits-all" approach. The knowledge of mechanisms responsible for MYC deregulation in MM enables identification of vulnerabilities and therapeutic targets in MYC-driven tumors. MYC can be targeted directly or indirectly, by interacting with several of its functions in cancer. Several such therapeutic strategies are evaluated in clinical trials in MM. In this review, we describe the mechanism of MYC activation in MM, the role of MYC in cancer progression, and the therapeutic options to targeting MYC.
Collapse
Affiliation(s)
| | - C Roche-Lestienne
- IRCL, INSERM UMR-S1172, Univ. Lille, Lille, France.,Institute of Medical Genetics, Univ. Lille, CHU, Lille, France
| | - I M Ghobrial
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - T Facon
- Department of Hematology, Univ. Lille,, CHU, Lille, France
| | - B Quesnel
- IRCL, INSERM UMR-S1172, Univ. Lille, Lille, France.,Department of Hematology, Univ. Lille,, CHU, Lille, France
| | - S Manier
- IRCL, INSERM UMR-S1172, Univ. Lille, Lille, France. .,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA. .,Department of Hematology, Univ. Lille,, CHU, Lille, France.
| |
Collapse
|
347
|
Abramson HN. Kinase inhibitors as potential agents in the treatment of multiple myeloma. Oncotarget 2018; 7:81926-81968. [PMID: 27655636 PMCID: PMC5348443 DOI: 10.18632/oncotarget.10745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Recent years have witnessed a dramatic increase in the number of therapeutic options available for the treatment of multiple myeloma (MM) - from immunomodulating agents to proteasome inhibitors to histone deacetylase (HDAC) inhibitors and, most recently, monoclonal antibodies. Used in conjunction with autologous hematopoietic stem cell transplantation, these modalities have nearly doubled the disease's five-year survival rate over the last three decades to about 50%. In spite of these advances, MM still is considered incurable as resistance and relapse are common. While small molecule protein kinase inhibitors have made inroads in the therapy of a number of cancers, to date their application to MM has been less than successful. Focusing on MM, this review examines the roles played by a number of kinases in driving the malignant state and the rationale for target development in the design of a number of kinase inhibitors that have demonstrated anti-myeloma activity in both in vitro and in vivo xenograph models, as well as those that have entered clinical trials. Among the targets and their inhibitors examined are receptor and non-receptor tyrosine kinases, cell cycle control kinases, the PI3K/AKT/mTOR pathway kinases, protein kinase C, mitogen-activated protein kinase, glycogen synthase kinase, casein kinase, integrin-linked kinase, sphingosine kinase, and kinases involved in the unfolded protein response.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
348
|
Pawlyn C, Loehr A, Ashby C, Tytarenko R, Deshpande S, Sun J, Fedorchak K, Mughal T, Davies FE, Walker BA, Morgan GJ. Loss of heterozygosity as a marker of homologous repair deficiency in multiple myeloma: a role for PARP inhibition? Leukemia 2018; 32:1561-1566. [PMID: 29467487 PMCID: PMC6035152 DOI: 10.1038/s41375-018-0017-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 10/31/2017] [Accepted: 12/15/2017] [Indexed: 02/07/2023]
Abstract
PARP inhibitors can induce synthetic lethality in tumors characterized by homologous recombination deficiency (HRD), which can be detected by evaluating genome-wide loss of heterozygosity (LOH). Multiple myeloma (MM) is a genetically unstable tumor and we hypothesized that HRD-related LOH (HRD-LOH) could be detected in patient samples, supporting a potential role for PARP inhibition in MM. Using results from targeted next-generation sequencing studies (FoundationOne® Heme), we analyzed HRD-LOH in patients at all disease stages (MGUS (n = 7), smoldering MM (SMM, n = 30), newly diagnosed MM (NDMM, n = 71), treated MM (TRMM, n = 64), and relapsed MM (RLMM, n = 234)) using an algorithm to identify HRD-LOH segments. We demonstrated HRD-LOH in MM samples, increasing as disease progresses. The extent of genomic HRD-LOH correlated with high-risk disease markers. Outcome of RLMM patients, the biggest clinical group, was analyzed and patients with HRD-LOH above the third quartile (≥5% HRD-LOH) had significantly worse progression-free and overall survival than those with lower levels (p < 0.001). Mutations in key homologous recombination genes account for some, but not all, of the cases with an excess of HRD-LOH. These data support the further evaluation of PARP inhibitors in MM patients, particularly in the relapsed setting with a high unmet need for new treatments.
Collapse
Affiliation(s)
| | | | - Cody Ashby
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ruslana Tytarenko
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shayu Deshpande
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - James Sun
- Foundation Medicine, Cambridge, MA, USA
| | | | | | - Faith E Davies
- 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
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
349
|
Tan D, Lee JH, Chen W, Shimizu K, Hou J, Suzuki K, Nawarawong W, Huang SY, Sang Chim C, Kim K, Kumar L, Malhotra P, Chng WJ, Durie B. Recent advances in the management of multiple myeloma: clinical impact based on resource-stratification. Consensus statement of the Asian Myeloma Network at the 16th international myeloma workshop. Leuk Lymphoma 2018; 59:2305-2317. [PMID: 29390932 DOI: 10.1080/10428194.2018.1427858] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Predicated on our improved understanding of the disease biology, we have seen remarkable advances in the management of multiple myeloma over the past few years. Recently approved drugs have radically transformed the treatment paradigm and improved survivals of myeloma patients. The progress has necessitated revision of the diagnostic criteria, risk-stratification and response definition. The huge disparities in economy, healthcare infrastructure and access to novel drugs among different Asian countries will hinder the delivery of optimum myeloma care to patients managed in resource-constrained environments. In the light of the tremendous recent changes and evolution in myeloma management, it is timely that the resource-stratified guidelines from the Asian Myeloma Network be revised to provide updated recommendations for Asia physicians practicing under various healthcare reimbursement systems. This review will highlight the most recent advances and our recommendations on how they could be integrated in both resource-abundant and resource-constrained facilities.
Collapse
Affiliation(s)
- Daryl Tan
- a Raffles Cancer Center , Raffles Hospital , Singapore.,b Department of Hematology , Singapore General Hospital , Singapore
| | - Jae Hoon Lee
- c Gil Hospital, Gachon University , Incheon , South Korea
| | - Wenming Chen
- d Beijing Chaoyang Hospital, Capital Medical University , Beijing , China
| | - Kazuyuki Shimizu
- e Higashi Nagoya National Hospital , National Hospital Organization , Nagoya , Japan
| | - Jian Hou
- f Department of Haematology , Changzheng Hospital, The Second Military Medical University , Shanghai , China
| | - Kenshi Suzuki
- g Department of Hematology , Japanese Red Cross Medical Center , Tokyo , Japan
| | | | | | - Chor Sang Chim
- j Queen Mary Hospital, University of Hong Kong , Hong Kong , China
| | - Kihyun Kim
- k Samsung Medical Center , Sungkyunkwan University , Seoul , South Korea
| | - Lalit Kumar
- l Department of Medical Oncology , Institute Rotary Cancer Hospital, All India Institute of Medical Sciences , New Delhi , India
| | - Pankaj Malhotra
- m Department of Internal Medicine , Postgraduate Institute of Medical Education and Research , Chandigarh , India
| | - Wee Joo Chng
- n Cancer Science Institute of Singapore , National University of Singapore , Singapore.,o Department of Haematology-Oncology , National University Cancer Institute of Singapore National University Health System , Singapore
| | - Brian Durie
- p Cedars-Sinai Comprehensive Cancer Center , Los Angeles , CA , USA
| | | |
Collapse
|
350
|
Walker BA. Whole Exome Sequencing in Multiple Myeloma to Identify Somatic Single Nucleotide Variants and Key Translocations Involving Immunoglobulin Loci and MYC. Methods Mol Biol 2018; 1792:71-95. [PMID: 29797253 DOI: 10.1007/978-1-4939-7865-6_6] [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: 12/12/2022]
Abstract
Multiple myeloma is a malignancy of terminally differentiated plasma cells in the bone marrow. These plasma cells produce high levels of immunoglobulin which cause end-organ damage. Rearrangements within the immunoglobulin loci are a physiological part of B cell development, but these DNA level double-strand breaks may result in interchromosomal translocations. There are five main translocations involving the Ig loci: t(4;14) 12%, t(6;14) 1%, t(11;14) 15%, t(14;16) 3%, and t(14;20) 2%. These are primary events, found in all cells within the tumor clone and are associated with different prognosis. The t(4;14), t(14;16), and t(14;20) are associated with a poor prognosis, whereas the others are associated with a more favorable prognosis. Rearrangements at the MYC locus are also associated with a poor prognosis and increased expression of MYC. MYC rearrangements are frequent (25%) and involve interchromosomal translocations involving Ig loci or other partners, but also include intrachromosomal inversions, duplications and deletions. As such, the Ig and MYC loci are key players in the myeloma genome and including these in any genomic studies is key to understanding the relationship with other abnormalities. We have designed a custom capture of the Ig and MYC loci which can be added to exome or targeted captures to inform on these key events. This saves on performing additional tests to determine these events, which are generally mandatory for any genetic investigations in myeloma. This custom capture is also relevant to other B cell malignancies where MYC and Ig translocations occur.
Collapse
Affiliation(s)
- Brian A Walker
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|