51
|
Sundararaj S, Seneviratne S, Williams SJ, Enders A, Casarotto MG. Structural determinants of the IRF4/DNA homodimeric complex. Nucleic Acids Res 2021; 49:2255-2265. [PMID: 33533913 PMCID: PMC7913761 DOI: 10.1093/nar/gkaa1287] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/22/2020] [Accepted: 02/01/2021] [Indexed: 11/15/2022] Open
Abstract
Interferon regulatory factor 4 (IRF4) is a key transcription factor (TF) in the regulation of immune cells, including B and T cells. It acts by binding DNA as both a homodimer and, in conjunction with other TFs, as a heterodimer. The choice of homo and heterodimeric/ DNA interactions is a critical aspect in the control of the transcriptional program and cell fate outcome. To characterize the nature of this interaction in the homodimeric complex, we have determined the crystal structure of the IRF4/ISRE homodimeric complex. We show that the complex formation is aided by a substantial DNA deformation with co-operative binding achieved exclusively through protein–DNA contact. This markedly contrasts with the heterodimeric form where DNA bound IRF4 is shown to physically interact with PU.1 TF to engage EICE1. We also show that the hotspot residues (Arg98, Cys99 and Asn102) contact both consensus and non-consensus sequences with the L1 loop exhibiting marked flexibility. Additionally, we identified that IRF4L116R, a mutant associated with chronic lymphocytic leukemia, binds more robustly to DNA thereby providing a rationale for the observed gain of function. Together, we demonstrate key structural differences between IRF4 homo and heterodimeric complexes, thereby providing molecular insights into IRF4-mediated transcriptional regulation.
Collapse
Affiliation(s)
- Srinivasan Sundararaj
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra 2600, Australia
| | - Sandali Seneviratne
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra 2600, Australia
| | - Simon J Williams
- Research School of Biology, Australian National University, Canberra 2600, Australia
| | - Anselm Enders
- Department of Immunology, John Curtin School of Medical Research, Australian National University, Canberra 2600, Australia.,Center for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Canberra 2600, Australia
| | - Marco G Casarotto
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra 2600, Australia
| |
Collapse
|
52
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
53
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
54
|
Analysis of Intratumoral Heterogeneity in Myelodysplastic Syndromes with Isolated del(5q) Using a Single Cell Approach. Cancers (Basel) 2021; 13:cancers13040841. [PMID: 33671317 PMCID: PMC7922695 DOI: 10.3390/cancers13040841] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic stem cell malignancies characterized by ineffective differentiation of one or more bone marrow cell lineages. Only 50% of patients with de novo MDS will be found to have cytogenetic abnormalities, of which del(5q) is the most common. In 10% of MDS cases, del(5q) is found as a sole abnormality. In this work, a single cell approach was used to analyze intratumoral heterogeneity in four patients with MDS with isolated del(5q). We were able to observe that an ancestral event in one patient can appear as a secondary hit in another one, thus reflecting the high intratumoral heterogeneity in MDS with isolated del(5q) and the importance of patient-specific molecular characterization. Abstract Myelodysplastic syndromes (MDS) are a heterogeneous group of hematological diseases. Among them, the most well characterized subtype is MDS with isolated chromosome 5q deletion (MDS del(5q)), which is the only one defined by a cytogenetic abnormality that makes these patients candidates to be treated with lenalidomide. During the last decade, single cell (SC) analysis has emerged as a powerful tool to decipher clonal architecture and to further understand cancer and other diseases at higher resolution level compared to bulk sequencing techniques. In this study, a SC approach was used to analyze intratumoral heterogeneity in four patients with MDS del(5q). Single CD34+CD117+CD45+CD19- bone marrow hematopoietic stem progenitor cells were isolated using the C1 system (Fluidigm) from diagnosis or before receiving any treatment and from available follow-up samples. Selected somatic alterations were further analyzed in SC by high-throughput qPCR (Biomark HD, Fluidigm) using specific TaqMan assays. A median of 175 cells per sample were analyzed. Inferred clonal architectures were relatively simple and either linear or branching. Similar to previous studies based on bulk sequencing to infer clonal architecture, we were able to observe that an ancestral event in one patient can appear as a secondary hit in another one, thus reflecting the high intratumoral heterogeneity in MDS del(5q) and the importance of patient-specific molecular characterization.
Collapse
|
55
|
Jackson GH, Pawlyn C, Cairns DA, de Tute RM, Hockaday A, Collett C, Jones JR, Kishore B, Garg M, Williams CD, Karunanithi K, Lindsay J, Rocci A, Snowden JA, Jenner MW, Cook G, Russell NH, Drayson MT, Gregory WM, Kaiser MF, Owen RG, Davies FE, Morgan GJ, the UK NCRI Haemato-oncology Clinical Studies Group. Carfilzomib, lenalidomide, dexamethasone, and cyclophosphamide (KRdc) as induction therapy for transplant-eligible, newly diagnosed multiple myeloma patients (Myeloma XI+): Interim analysis of an open-label randomised controlled trial. PLoS Med 2021; 18:e1003454. [PMID: 33428632 PMCID: PMC7799846 DOI: 10.1371/journal.pmed.1003454] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 11/23/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Carfilzomib is a second-generation irreversible proteasome inhibitor that is efficacious in the treatment of myeloma and carries less risk of peripheral neuropathy than first-generation proteasome inhibitors, making it more amenable to combination therapy. METHODS AND FINDINGS The Myeloma XI+ trial recruited patients from 88 sites across the UK between 5 December 2013 and 20 April 2016. Patients with newly diagnosed multiple myeloma eligible for transplantation were randomly assigned to receive the combination carfilzomib, lenalidomide, dexamethasone, and cyclophosphamide (KRdc) or a triplet of lenalidomide, dexamethasone, and cyclophosphamide (Rdc) or thalidomide, dexamethasone, and cyclophosphamide (Tdc). All patients were planned to receive an autologous stem cell transplantation (ASCT) prior to a randomisation between lenalidomide maintenance and observation. Eligible patients were aged over 18 years and had symptomatic myeloma. The co-primary endpoints for the study were progression-free survival (PFS) and overall survival (OS) for KRdc versus the Tdc/Rdc control group by intention to treat. PFS, response, and safety outcomes are reported following a planned interim analysis. The trial is registered (ISRCTN49407852) and has completed recruitment. In total, 1,056 patients (median age 61 years, range 33 to 75, 39.1% female) underwent induction randomisation to KRdc (n = 526) or control (Tdc/Rdc, n = 530). After a median follow-up of 34.5 months, KRdc was associated with a significantly longer PFS than the triplet control group (hazard ratio 0.63, 95% CI 0.51-0.76). The median PFS for patients receiving KRdc is not yet estimable, versus 36.2 months for the triplet control group (p < 0.001). Improved PFS was consistent across subgroups of patients including those with genetically high-risk disease. At the end of induction, the percentage of patients achieving at least a very good partial response was 82.3% in the KRdc group versus 58.9% in the control group (odds ratio 4.35, 95% CI 3.19-5.94, p < 0.001). Minimal residual disease negativity (cutoff 4 × 10-5 bone marrow leucocytes) was achieved in 55% of patients tested in the KRdc group at the end of induction, increasing to 75% of those tested after ASCT. The most common adverse events were haematological, with a low incidence of cardiac events. The trial continues to follow up patients to the co-primary endpoint of OS and for planned long-term follow-up analysis. Limitations of the study include a lack of blinding to treatment regimen and that the triplet control regimen did not include a proteasome inhibitor for all patients, which would be considered a current standard of care in many parts of the world. CONCLUSIONS The KRdc combination was well tolerated and was associated with both an increased percentage of patients achieving at least a very good partial response and a significant PFS benefit compared to immunomodulatory-agent-based triplet therapy. TRIAL REGISTRATION ClinicalTrials.gov ISRCTN49407852.
Collapse
Affiliation(s)
- Graham H. Jackson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Charlotte Pawlyn
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - David A. Cairns
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Ruth M. de Tute
- Haematological Malignancy Diagnostic Service, St James’s University Hospital, Leeds, United Kingdom
| | - Anna Hockaday
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Corinne Collett
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - John R. Jones
- Kings College Hospital NHS Foundation Trust, London, United Kingdom
| | - Bhuvan Kishore
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Mamta Garg
- Leicester Royal Infirmary, Leicester, United Kingdom
| | - Cathy D. Williams
- Centre for Clinical Haematology, Nottingham University Hospital, Nottingham, United Kingdom
| | - Kamaraj Karunanithi
- University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, United Kingdom
| | - Jindriska Lindsay
- East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom
| | - Alberto Rocci
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - John A. Snowden
- Royal Hallamshire Hospital, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Matthew W. Jenner
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Gordon Cook
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
- Section of Experimental Haematology, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Nigel H. Russell
- Centre for Clinical Haematology, Nottingham University Hospital, Nottingham, United Kingdom
| | - Mark T. Drayson
- Clinical Immunology Service, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Walter M. Gregory
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, United Kingdom
| | - Martin F. Kaiser
- The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
| | - Roger G. Owen
- Haematological Malignancy Diagnostic Service, St James’s University Hospital, Leeds, United Kingdom
| | - Faith E. Davies
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, United States of America
| | - Gareth J. Morgan
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, United States of America
| | | |
Collapse
|
56
|
Fraga de Andrade I, Mehta C, Bresnick EH. Post-transcriptional control of cellular differentiation by the RNA exosome complex. Nucleic Acids Res 2020; 48:11913-11928. [PMID: 33119769 PMCID: PMC7708067 DOI: 10.1093/nar/gkaa883] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/21/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022] Open
Abstract
Given the complexity of intracellular RNA ensembles and vast phenotypic remodeling intrinsic to cellular differentiation, it is instructive to consider the role of RNA regulatory machinery in controlling differentiation. Dynamic post-transcriptional regulation of protein-coding and non-coding transcripts is vital for establishing and maintaining proteomes that enable or oppose differentiation. By contrast to extensively studied transcriptional mechanisms governing differentiation, many questions remain unanswered regarding the involvement of post-transcriptional mechanisms. Through its catalytic activity to selectively process or degrade RNAs, the RNA exosome complex dictates the levels of RNAs comprising multiple RNA classes, thereby regulating chromatin structure, gene expression and differentiation. Although the RNA exosome would be expected to control diverse biological processes, studies to elucidate its biological functions and how it integrates into, or functions in parallel with, cell type-specific transcriptional mechanisms are in their infancy. Mechanistic analyses have demonstrated that the RNA exosome confers expression of a differentiation regulatory receptor tyrosine kinase, downregulates the telomerase RNA component TERC, confers genomic stability and promotes DNA repair, which have considerable physiological and pathological implications. In this review, we address how a broadly operational RNA regulatory complex interfaces with cell type-specific machinery to control cellular differentiation.
Collapse
Affiliation(s)
- Isabela Fraga de Andrade
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, 4009 WIMR, Madison, WI 53705, USA
| | - Charu Mehta
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, 4009 WIMR, Madison, WI 53705, USA
| | - Emery H Bresnick
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, 4009 WIMR, Madison, WI 53705, USA
| |
Collapse
|
57
|
Xiang X, He Q, Ou Y, Wang W, Wu Y. Efficacy and Safety of CAR-Modified T Cell Therapy in Patients with Relapsed or Refractory Multiple Myeloma: A Meta-Analysis of Prospective Clinical Trials. Front Pharmacol 2020; 11:544754. [PMID: 33343342 PMCID: PMC7744881 DOI: 10.3389/fphar.2020.544754] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 10/16/2020] [Indexed: 02/05/2023] Open
Abstract
Background: In recent years, chimeric antigen receptor-modified T (CAR-T) cell therapy for B-cell leukemia and lymphoma has shown high clinical efficacy. Similar CAR-T clinical trials have also been carried out in patients with refractory/relapsed multiple myeloma (RRMM). However, no systematic review has evaluated the efficacy and safety of CAR-T cell therapy in RRMM. The purpose of this study was to fill this literature gap. Methods: Eligible studies were searched in PUBMED, EMBASE, the Cochrane Central Register of Controlled Trials (CENTRAL), CNKI, and WanFang from data inception to December 2019. For efficacy assessment, the overall response rate (ORR), minimal residual disease (MRD) negativity rate, strict complete response (sCR), complete response (CR), very good partial response (VGPR), and partial response (PR) were calculated. The incidence of any grade cytokine release syndrome (CRS) and grade ≥3 adverse events (AEs) were calculated for safety analysis. The effect estimates were then pooled using an inverse variance method. Results: Overall, 27 studies involving 497 patients were included in this meta-analysis. The pooled ORR and MRD negativity rate were 89% (95% Cl: 83-94%) and 81% (95% Cl: 67-91%), respectively. The pooled sCR, CR, VGPR, and PR were 14% (95% Cl: 5-27%), 13% (95% Cl: 4-26%), 23% (95% Cl: 14-33%), and 15% (95% Cl: 10-21%), respectively. Subgroup analyses of ORR by age, proportion of previous autologous stem cell transplantation (ASCT), and target selection of CAR-T cells revealed that age ≤ 55 years (≤55 years vs. > 55 years, p = 0.0081), prior ASCT ≤70% (≤70% vs. > 70%, p = 0.035), and bispecific CAR-T cells (dual B-cell maturation antigen (BCMA)/BCMA + CD19 vs specific BCMA, p = 0.0329) associated with higher ORR in patients. Subgroup analyses of remission depth by target selection suggested that more patients achieved a better response than VGPR with dual BCMA/BCMA + CD19 CAR-T cells compared to specific BCMA targeting (p = 0.0061). In terms of safety, the pooled incidence of any grade and grade ≥ 3 CRS was 76% (95% CL: 63-87%) and 11% (95% CL: 6-17%). The most common grade ≥ 3 AEs were hematologic toxic effects. Conclusion: In heavily treated patients, CAR-T therapy associates with promising responses and tolerable AEs, as well as CRS in RRMM. However, additional information regarding the durability of CAR-T cell therapy, as well as further randomized controlled trials, is needed.
Collapse
Affiliation(s)
- Xinrong Xiang
- Hematology Research Laboratory, West China Hospital, Department of Hematology, Sichuan University, Chengdu, China
| | - Qiao He
- Chinese Evidence-based Medicine Center and Cochrane China Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Ou
- Hematology Research Laboratory, West China Hospital, Department of Hematology, Sichuan University, Chengdu, China
| | - Wen Wang
- Chinese Evidence-based Medicine Center and Cochrane China Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Wu
- Hematology Research Laboratory, West China Hospital, Department of Hematology, Sichuan University, Chengdu, China
| |
Collapse
|
58
|
Bolli N, Martinelli G, Cerchione C. The molecular pathogenesis of multiple myeloma. Hematol Rep 2020; 12:9054. [PMID: 33408844 PMCID: PMC7772755 DOI: 10.4081/hr.2020.9054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/29/2022] Open
Abstract
Multiple Myeloma (MM) is characterized by uncontrolled proliferation and accumulation of clonal plasma cells within the bone marrow. However, the cell of origin is a B-lymphocyte acquiring aberrant genomic events in the germinal center of a lymph node as off-target events during somatichypermutation and class-switch recombination driven by activation-induced-deaminase. Whether pre-germinal center events are also required for transformation, and which additional events are required for disease progression is still matter of debate. As early treatment in asymptomatic phases is gaining traction in the clinic, a better understanding of the molecular pathogenesis of myeloma progression would allow stratification of patients based on their risk of progression, thus rationalizing efficacy and cost of clinical interventions. In this review, we will discuss the development of MM, from the cell of origin through asymptomatic stages such as monoclonal gammopathy of undetermined significance and smoldering MM, to the development of symptomatic disease. We will explain the genetic heterogeneity of MM, one of the major drivers of disease recurrence. In this context, moreover, we will propose how this knowledge may influence future diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Niccolò Bolli
- Department of Oncology and Hemato- Oncology, University of Milan, Italy
- Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Martinelli
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
59
|
Zhu X, Li S, Xu B, Luo H. Cancer evolution: A means by which tumors evade treatment. Biomed Pharmacother 2020; 133:111016. [PMID: 33246226 DOI: 10.1016/j.biopha.2020.111016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
Although various methods have been tried to study and treat cancer, the cancer remains a major challenge for human medicine today. One important reason for this is the presence of cancer evolution. Cancer evolution is a process in which tumor cells adapt to the external environment, which can suppress the human immune system's ability to recognize and attack tumors, and also reduce the reproducibility of cancer research. Among them, heterogeneity of the tumor provides intrinsic motivation for this process. Recently, with the development of related technologies such as liquid biopsy, more and more knowledge about cancer evolution has been gained and interest in this topic has also increased. Therefore, starting from the causes of tumorigenesis, this paper introduces several tumorigenesis processes and pathways, as well as treatment options for different targets.
Collapse
Affiliation(s)
- Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Shi Li
- Guangdong Key Laboratory of Urogenital Tumor Systems and Synthetic Biology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China; Shenzhen Key Laboratory of Genitourinary Tumor, Translational Medicine Institute of Shenzhen, The Second People's Hospital of Shenzhen, Shenzhen, China; College of Bioengineering, Chongqing University, Chongqing, China
| | - Bairui Xu
- The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjian, China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjian, China.
| |
Collapse
|
60
|
Changing paradigms in diagnosis and treatment of monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM). Leukemia 2020; 34:3111-3125. [PMID: 33046818 DOI: 10.1038/s41375-020-01051-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/18/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
Multiple myeloma (MM) is a highly heterogenous disease that exists along a continuous disease spectrum starting with premalignant conditions monoclonal gammopathy of undetermined significance (MGUS) and smoldering multiple myeloma (SMM) that inevitably precede MM. Over the past two decades, significant progress has been made in the genetic characterization and risk stratification of precursor plasma cell disorders. Indeed, the clinical introduction of highly effective and well-tolerated drugs begs the question: would earlier therapeutic intervention with novel therapies in MGUS and SMM patients alter natural history, providing a potential curative option? In this review, we discuss the epidemiology of MGUS and SMM and current models for risk stratification that predict MGUS and SMM progression to MM. We further discuss genetic heterogeneity and clonal evolution in MM and the interplay between tumor cells and the bone marrow (BM) microenvironment. Finally, we provide an overview of the current recommendations for the management of MGUS and SMM and discuss the open controversies in the field in light of promising results from early intervention clinical trials.
Collapse
|
61
|
Impact of last lenalidomide dose, duration, and IMiD-free interval in patients with myeloma treated with pomalidomide/dexamethasone. Blood Adv 2020; 3:4095-4103. [PMID: 31821457 DOI: 10.1182/bloodadvances.2019000539] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022] Open
Abstract
To gain insights into the characteristics of clinical resistance to lenalidomide, we evaluated the outcomes of 147 consecutive patients with multiple myeloma (MM) homogeneously treated with immunomodulatory imide drugs (IMiDs) pomalidomide and dexamethasone (Pd) for relapsed and/or refractory MM (median, 3 prior lines of treatment). We focused our analysis on the effect of the lenalidomide dose at which resistance was developed, the duration of lenalidomide exposure, and lenalidomide-free interval. On intent to treat, 33% of patients achieved ≥partial remission (PR) with Pd. When Pd was given immediately after lenalidomide, ≥PR was 32% (vs 37% after bortezomib). The response rates were similar for patients that received 5 to 15 mg vs 25 mg of lenalidomide (38.5% vs 30.5%, P = .329). Response rates were higher for patients that had received at least 12 months of lenalidomide (44% vs 27%) and for those with ≥18 months from last lenalidomide dose to pomalidomide dose (65% vs 23%). Median progression-free survival (PFS) and overall survival (OS) were 5 and 12.1 months, respectively, which was similar for patients who received lenalidomide, bortezomib or other regimens just before Pd and similar for patients who were receiving different doses of lenalidomide. IMiD-free interval ≥18 months was associated with longer PFS (10.3 vs 3.9 months, P = .003) and OS (27.1 vs 9.3, P = .008) as well as duration of last lenalidomide therapy ≥12 months (PFS: 7.8 vs 3.2, P = .023; OS: 16.5 vs 7.9, P = .005) even after adjustment for the number of prior therapies, duration of disease, and last lenalidomide dose.
Collapse
|
62
|
Williams LS, Caro J, Razzo B, Boyle EM, Morgan GJ. Deep sequencing as an approach to understanding the complexity and improving the treatment of multiple myeloma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1792285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Louis S. Williams
- Department of Hematology & Medical Oncology, NYU Langone Medical Center, New York, NY, USA
| | - Jessica Caro
- Department of Hematology & Medical Oncology, NYU Langone Medical Center, New York, NY, USA
| | - Beatrice Razzo
- Department of Internal Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Eileen M. Boyle
- Department of Hematology & Medical Oncology, Multiple Myeloma Research Program, NYU Langone Medical Center, New York, NY, USA
| | - Gareth J. Morgan
- Department of Hematology & Medical Oncology, Multiple Myeloma Research Program, NYU Langone Medical Center, New York, NY, USA
| |
Collapse
|
63
|
Foltz SM, Gao Q, Yoon CJ, Sun H, Yao L, Li Y, Jayasinghe RG, Cao S, King J, Kohnen DR, Fiala MA, Ding L, Vij R. Evolution and structure of clinically relevant gene fusions in multiple myeloma. Nat Commun 2020; 11:2666. [PMID: 32471990 PMCID: PMC7260243 DOI: 10.1038/s41467-020-16434-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 04/27/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma is a plasma cell blood cancer with frequent chromosomal translocations leading to gene fusions. To determine the clinical relevance of fusion events, we detect gene fusions from a cohort of 742 patients from the Multiple Myeloma Research Foundation CoMMpass Study. Patients with multiple clinic visits enable us to track tumor and fusion evolution, and cases with matching peripheral blood and bone marrow samples allow us to evaluate the concordance of fusion calls in patients with high tumor burden. We examine the joint upregulation of WHSC1 and FGFR3 in samples with t(4;14)-related fusions, and we illustrate a method for detecting fusions from single cell RNA-seq. We report fusions at MYC and a neighboring gene, PVT1, which are related to MYC translocations and associated with divergent progression-free survival patterns. Finally, we find that 4% of patients may be eligible for targeted fusion therapies, including three with an NTRK1 fusion.
Collapse
Affiliation(s)
- Steven M Foltz
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Qingsong Gao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Christopher J Yoon
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Hua Sun
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Lijun Yao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Reyka G Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Justin King
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Daniel R Kohnen
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Mark A Fiala
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA.
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| | - Ravi Vij
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63110, USA.
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63110, USA.
| |
Collapse
|
64
|
Eisfeld C, Eßeling E, Wullenkord R, Khandanpour C, Reusch J, Mikesch JH, Reicherts C, Kerkhoff A, Schliemann C, Kessler T, Mesters RM, Berdel WE, Lenz G, Stelljes M. Long-term survival and polyclonal immunoglobulin reconstitution after allogeneic stem cell transplantation in multiple myeloma. Ann Hematol 2020; 99:1907-1915. [PMID: 32444892 PMCID: PMC7340674 DOI: 10.1007/s00277-020-04068-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/28/2020] [Indexed: 01/19/2023]
Abstract
Despite significant progress made in the treatment of patients with multiple myeloma (MM) in the last decade, for patients with early relapse or rapidly progressing high-risk disease, allogeneic hematopoietic stem cell transplantation (SCT) might be an option leading to long-term survival. Here, we retrospectively analyzed the outcomes of 90 MM patients who received allogeneic SCT in our center between 1999 and 2017. We specifically assessed the association of impaired humoral immune reconstitution, referred to as immunoparesis, and post-transplant survival. Sixty-four patients received allogeneic SCT in relapse following 2–7 lines of therapy; 26 patients received upfront tandem autologous-allogeneic SCT. With a median follow-up of 76 months, OS and PFS were 52.6% (95% CI 42.9–64.3) and 36.4% (95% CI 27.6–47.9) at 2 years and 38.6% (95% CI 29.2–51.1) and 25.3% (95% CI 17.5–36.4) at 5 years, respectively. Receiving more than two therapy lines prior to transplantation was an independent risk factor for OS (HR 3.68, 95% CI 2.02–6.70) and PFS (HR 3.69, 95% CI 2.09–6.50). In a landmark analysis at day 200, prolonged immunoparesis was associated with reduced OS (HR 3.22, 95% CI 1.14–9.11). Allogeneic stem cell transplantation offers an additional treatment element that may lead to long-term remission in selected patients with poor prognosis, probably exploiting graft-versus-myeloma effects. Immunoparesis could potentially serve as an indicator for impaired survival following allogeneic transplantation, an observation to be further studied prospectively.
Collapse
Affiliation(s)
- Christine Eisfeld
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Eva Eßeling
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Ramona Wullenkord
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Cyrus Khandanpour
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Julia Reusch
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | | | | - Andrea Kerkhoff
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | | | - Torsten Kessler
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Rolf M Mesters
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Matthias Stelljes
- Department of Medicine A, University Hospital Münster, Münster, Germany.
| |
Collapse
|
65
|
Ferreira B, Caetano J, Barahona F, Lopes R, Carneiro E, Costa-Silva B, João C. Liquid biopsies for multiple myeloma in a time of precision medicine. J Mol Med (Berl) 2020; 98:513-525. [PMID: 32246161 PMCID: PMC7198642 DOI: 10.1007/s00109-020-01897-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/06/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Multiple myeloma (MM) is a challenging, progressive, and highly heterogeneous hematological malignancy. MM is characterized by multifocal proliferation of neoplastic plasma cells in the bone marrow (BM) and sometimes in extramedullary organs. Despite the availability of novel drugs and the longer median overall survival, some patients survive more than 10 years while others die rapidly. This heterogeneity is mainly driven by biological characteristics of MM cells, including genetic abnormalities. Disease progressions are mainly due to the inability of drugs to overcome refractory disease and inevitable drug-resistant relapse. In clinical practice, a bone marrow biopsy, mostly performed in one site, is still used to access the genetics of MM. However, BM biopsy use is limited by its invasive nature and by often not accurately reflecting the mutational profile of MM. Recent insights into the genetic landscape of MM provide a valuable opportunity to implement precision medicine approaches aiming to enable better patient profiling and selection of targeted therapies. In this review, we explore the use of the emerging field of liquid biopsies in myeloma patients considering current unmet medical needs, such as assessing the dynamic mutational landscape of myeloma, early predictors of treatment response, and a less invasive response monitoring.
Collapse
Affiliation(s)
- Bruna Ferreira
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Joana Caetano
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Filipa Barahona
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Raquel Lopes
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Emilie Carneiro
- Myeloma and Lymphoma Research Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cristina João
- Hemato-Oncology Unit, Myeloma and Lymphoma Research Programme, Nova Medical School, Champalimaud Centre for the Unknown, Lisbon, Portugal.
| |
Collapse
|
66
|
Maclachlan K, Diamond B, Maura F, Hillengass J, Turesson I, Landgren CO, Kazandjian D. Second malignancies in multiple myeloma; emerging patterns and future directions. Best Pract Res Clin Haematol 2020; 33:101144. [PMID: 32139010 PMCID: PMC7544243 DOI: 10.1016/j.beha.2020.101144] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 12/20/2022]
Abstract
The changing landscape of treatment options for multiple myeloma has led to a higher proportion of patients achieving deep, long-lasting responses to therapy. With the associated improvement in overall survival, the development of subsequent second malignancies has become of increased significance. The risk of second malignancy after multiple myeloma is affected by a combination of patient-, disease- and therapy-related risk factors. This review discusses recent data refining our knowledge of these contributing factors, including current treatment modalities which increase risk (i.e. high-dose melphalan with autologous stem cell transplant and lenalidomide maintenance therapy). We highlight emerging data towards individualized risk- and response-adapted treatment strategies and discuss key areas requiring future research.
Collapse
Affiliation(s)
- Kylee Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Benjamin Diamond
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francesco Maura
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jens Hillengass
- Section of Multiple Myeloma, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ingemar Turesson
- Department of Hematology, Skane University Hospital, Malmo, Sweden
| | - C Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dickran Kazandjian
- Multiple Myeloma Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
67
|
Exon-4 Mutations in KRAS Affect MEK/ERK and PI3K/AKT Signaling in Human Multiple Myeloma Cell Lines. Cancers (Basel) 2020; 12:cancers12020455. [PMID: 32079091 PMCID: PMC7072554 DOI: 10.3390/cancers12020455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/04/2020] [Accepted: 02/13/2020] [Indexed: 01/12/2023] Open
Abstract
Approximately 20% of multiple myeloma (MM) cases harbor a point mutation in KRAS. However, there is still no final consent on whether KRAS-mutations are associated with disease outcome. Specifically, no data exist on whether KRAS-mutations have an impact on survival of MM patients at diagnosis in the era of novel agents. Direct blockade of KRAS for therapeutic purposes is mostly impossible, but recently a mutation-specific covalent inhibitor targeting KRASp.G12C entered into clinical trials. However, other KRAS hotspot-mutations exist in MM patients, including the less common exon-4 mutations. For the current study, the coding regions of KRAS were deep-sequenced in 80 newly diagnosed MM patients, uniformely treated with three cycles of bortezomib plus dexamethasone and cyclophosphamide (VCD)-induction, followed by high-dose chemotherapy and autologous stem cell transplantation. Moreover, the functional impact of KRASp.G12A and the exon-4 mutations p.A146T and p.A146V on different survival pathways was investigated. Specifically, KRASWT, KRASp.G12A, KRASp.A146T, and KRASp.A146V were overexpressed in HEK293 cells and the KRASWT MM cell lines JJN3 and OPM2 using lentiviral transduction and the Sleeping Beauty vector system. Even though KRAS-mutations were not correlated with survival, all KRAS-mutants were found capable of potentially activating MEK/ERK- and sustaining PI3K/AKT-signaling in MM cells.
Collapse
|
68
|
Furukawa Y, Kikuchi J. Molecular basis of clonal evolution in multiple myeloma. Int J Hematol 2020; 111:496-511. [DOI: 10.1007/s12185-020-02829-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
|
69
|
Friend N, Noll JE, Opperman KS, Clark KC, Mrozik KM, Vandyke K, Hewett DR, Zannettino ACW. GLIPR1 expression is reduced in multiple myeloma but is not a tumour suppressor in mice. PLoS One 2020; 15:e0228408. [PMID: 31995627 PMCID: PMC6988976 DOI: 10.1371/journal.pone.0228408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/14/2020] [Indexed: 01/08/2023] Open
Abstract
Multiple myeloma, a plasma cell malignancy, is a genetically heterogeneous disease and the genetic factors that contribute to its development and progression remain to be fully elucidated. The tumour suppressor gene GLIPR1 has previously been shown to be deleted in approximately 10% of myeloma patients, to inhibit the development of plasma cell tumours in ageing mice and to have reduced expression levels in the plasma cells of patients with light-chain amyloidosis, a myeloma-related malignancy. Therefore, we hypothesised that GLIPR1 may have tumour suppressor activity in multiple myeloma. In this study, we demonstrate that plasma cell expression of GLIPR1 is reduced in the majority of myeloma patients and Glipr1 expression is lost in the 5TGM1 murine myeloma cell line. However, overexpression of GLIPR1 in a human myeloma cell line did not affect cell proliferation in vitro. Similarly, re-expression of Glipr1 in 5TGM1 cells did not significantly reduce their in vitro proliferation or in vivo growth in C57BL/KaLwRij mice. In addition, using CRISPR-Cas9 genome editing, we generated C57BL/Glipr1-/- mice and showed that loss of Glipr1 in vivo did not affect normal haematopoiesis or the development of monoclonal plasma cell expansions in these mice up to one year of age. Taken together, our results suggest that GLIPR1 is unlikely to be a potent tumour suppressor in multiple myeloma. However, it remains possible that the down-regulation of GLIPR1 may cooperate with other genetic lesions to promote the development of myeloma.
Collapse
Affiliation(s)
- Natasha Friend
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Jacqueline E. Noll
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Khatora S. Opperman
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Kimberley C. Clark
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Krzysztof M. Mrozik
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Kate Vandyke
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Duncan R. Hewett
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Andrew C. W. Zannettino
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
- * E-mail:
| |
Collapse
|
70
|
Wang Z, Zhao Y, An Z, Li W. Molecular Links Between Angiogenesis and Neuroendocrine Phenotypes in Prostate Cancer Progression. Front Oncol 2020; 9:1491. [PMID: 32039001 PMCID: PMC6985539 DOI: 10.3389/fonc.2019.01491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/11/2019] [Indexed: 12/11/2022] Open
Abstract
As a common therapy for prostate cancer, androgen deprivation therapy (ADT) is effective for the majority of patients. However, prolonged ADT promotes drug resistance and progression to an aggressive variant with reduced androgen receptor signaling, so called neuroendocrine prostate cancer (NEPC). Until present, NEPC is still poorly understood, and lethal with no effective treatments. Elevated expression of neuroendocrine related markers and increased angiogenesis are two prominent phenotypes of NEPC, and both of them are positively associated with cancers progression. However, direct molecular links between the two phenotypes in NEPC and their mechanisms remain largely unclear. Their elucidation should substantially expand our knowledge in NEPC. This knowledge, in turn, would facilitate the development of effective NEPC treatments. We recently showed that a single critical pathway regulates both ADT-enhanced angiogenesis and elevated expression of neuroendocrine markers. This pathway consists of CREB1, EZH2, and TSP1. Here, we seek new insights to identify molecules common to pathways promoting angiogenesis and neuroendocrine phenotypes in prostate cancer. To this end, our focus is to summarize the literature on proteins reported to regulate both neuroendocrine marker expression and angiogenesis as potential molecular links. These proteins, often described in separate biological contexts or diseases, include AURKA and AURKB, CHGA, CREB1, EZH2, FOXA2, GRK3, HIF1, IL-6, MYCN, ONECUT2, p53, RET, and RB1. We also present the current efforts in prostate cancer or other diseases to target some of these proteins, which warrants testing for NEPC, given the urgent unmet need in treating this aggressive variant of prostate cancer.
Collapse
Affiliation(s)
- Zheng Wang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - Yicheng Zhao
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, United States
| | - Wenliang Li
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, United States
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, United States
| |
Collapse
|
71
|
Abstract
The evolutionarily conserved RNA exosome is a multisubunit ribonuclease complex that processes and/or degrades numerous RNAs. Recently, mutations in genes encoding both structural and catalytic subunits of the RNA exosome have been linked to human disease. Mutations in the structural exosome gene EXOSC2 cause a distinct syndrome that includes retinitis pigmentosa, hearing loss, and mild intellectual disability. In contrast, mutations in the structural exosome genes EXOSC3 and EXOSC8 cause pontocerebellar hypoplasia type 1b (PCH1b) and type 1c (PCH1c), respectively, which are related autosomal recessive, neurodegenerative diseases. In addition, mutations in the structural exosome gene EXOSC9 cause a PCH-like disease with cerebellar atrophy and spinal motor neuronopathy. Finally, mutations in the catalytic exosome gene DIS3 have been linked to multiple myeloma, a neoplasm of plasma B cells. How mutations in these RNA exosome genes lead to distinct, tissue-specific diseases is not currently well understood. In this chapter, we examine the role of the RNA exosome complex in human disease and discuss the mechanisms by which mutations in different exosome subunit genes could impair RNA exosome function and give rise to diverse diseases.
Collapse
Affiliation(s)
- Milo B Fasken
- Department of Biology, RRC 1021, Emory University, Atlanta, GA, USA.
| | - Derrick J Morton
- Department of Biology, RRC 1021, Emory University, Atlanta, GA, USA
| | - Emily G Kuiper
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephanie K Jones
- Department of Biology, RRC 1021, Emory University, Atlanta, GA, USA
- Genetics and Molecular Biology Graduate Program, Emory University, Atlanta, GA, USA
| | - Sara W Leung
- Department of Biology, RRC 1021, Emory University, Atlanta, GA, USA
| | - Anita H Corbett
- Department of Biology, RRC 1021, Emory University, Atlanta, GA, USA.
| |
Collapse
|
72
|
Wu C, Zhang L, Brockman QR, Zhan F, Chen L. Chimeric antigen receptor T cell therapies for multiple myeloma. J Hematol Oncol 2019; 12:120. [PMID: 31752943 PMCID: PMC6873434 DOI: 10.1186/s13045-019-0823-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy and remains incurable despite the advent of numerous new drugs such as proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. There is an unmet need to develop novel therapies for refractory/relapsed MM. In the past few years, chimeric antigen receptor (CAR)-modified T cell therapy for MM has shown promising efficacy in preclinical and clinical studies. Furthermore, the toxicities of CAR-T cell therapy are manageable. This article summarizes recent developments of CAR-T therapy in MM, focusing on promising targets, new technologies, and new research areas. Additionally, a comprehensive overview of antigen selection is presented along with preliminary results and future directions of CAR-T therapy development.
Collapse
Affiliation(s)
- Chao Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Lina Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Qierra R Brockman
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA.,Molecular Medicine Program, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Fenghuang Zhan
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
| |
Collapse
|
73
|
Stefka AT, Johnson D, Rosebeck S, Park JH, Nakamura Y, Jakubowiak AJ. Potent anti-myeloma activity of the TOPK inhibitor OTS514 in pre-clinical models. Cancer Med 2019; 9:324-334. [PMID: 31714026 PMCID: PMC6943155 DOI: 10.1002/cam4.2695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/30/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) continues to be considered incurable, necessitating new drug discovery. The mitotic kinase T‐LAK cell‐originated protein kinase/PDZ‐binding kinase (TOPK/PBK) is associated with proliferation of tumor cells, maintenance of cancer stem cells, and poor patient prognosis in many cancers. In this report, we demonstrate potent anti‐myeloma effects of the TOPK inhibitor OTS514 for the first time. OTS514 induces cell cycle arrest and apoptosis at nanomolar concentrations in a series of human myeloma cell lines (HMCL) and prevents outgrowth of a putative CD138+ stem cell population from MM patient‐derived peripheral blood mononuclear cells. In bone marrow cells from MM patients, OTS514 treatment exhibited preferential killing of the malignant CD138+ plasma cells compared with the CD138− compartment. In an aggressive mouse xenograft model, OTS964 given orally at 100 mg/kg 5 days per week was well tolerated and reduced tumor size by 48%‐81% compared to control depending on the initial graft size. FOXO3 and its transcriptional targets CDKN1A (p21) and CDKN1B (p27) were elevated and apoptosis was induced with OTS514 treatment of HMCLs. TOPK inhibition also induced loss of FOXM1 and disrupted AKT, p38 MAPK, and NF‐κB signaling. The effects of OTS514 were independent of p53 mutation or deletion status. Combination treatment of HMCLs with OTS514 and lenalidomide produced synergistic effects, providing a rationale for the evaluation of TOPK inhibition in existing myeloma treatment regimens.
Collapse
Affiliation(s)
- Andrew T Stefka
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - David Johnson
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Shaun Rosebeck
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
74
|
Miura S, Huuki LA, Buturla T, Vu T, Gomez K, Kumar S. Computational enhancement of single-cell sequences for inferring tumor evolution. Bioinformatics 2019; 34:i917-i926. [PMID: 30423071 DOI: 10.1093/bioinformatics/bty571] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Motivation Tumor sequencing has entered an exciting phase with the advent of single-cell techniques that are revolutionizing the assessment of single nucleotide variation (SNV) at the highest cellular resolution. However, state-of-the-art single-cell sequencing technologies produce data with many missing bases (MBs) and incorrect base designations that lead to false-positive (FP) and false-negative (FN) detection of somatic mutations. While computational methods are available to make biological inferences in the presence of these errors, the accuracy of the imputed MBs and corrected FPs and FNs remains unknown. Results Using computer simulated datasets, we assessed the robustness performance of four existing methods (OncoNEM, SCG, SCITE and SiFit) and one new method (BEAM). BEAM is a Bayesian evolution-aware method that improves the quality of single-cell sequences by using the intrinsic evolutionary information in the single-cell data in a molecular phylogenetic framework. Overall, BEAM and SCITE performed the best. Most of the methods imputed MBs with high accuracy, but effective detection and correction of FPs and FNs is a challenge, especially for small datasets. Analysis of an empirical dataset shows that computational methods can improve both the quality of tumor single-cell sequences and their utility for biological inference. In conclusion, tumor cells descend from pre-existing cells, which creates evolutionary continuity in single-cell sequencing datasets. This information enables BEAM and other methods to correctly impute missing data and incorrect base assignments, but correction of FPs and FNs remains challenging when the number of SNVs sampled is small relative to the number of cells sequenced. Availability and implementation BEAM is available on the web at https://github.com/SayakaMiura/BEAM.
Collapse
Affiliation(s)
- Sayaka Miura
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.,Department of Biology, Temple University, Philadelphia, PA, USA
| | - Louise A Huuki
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.,Department of Biology, Temple University, Philadelphia, PA, USA
| | - Tiffany Buturla
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.,Department of Biology, Temple University, Philadelphia, PA, USA
| | - Tracy Vu
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.,Department of Biology, Temple University, Philadelphia, PA, USA
| | - Karen Gomez
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.,Department of Biology, Temple University, Philadelphia, PA, USA
| | - Sudhir Kumar
- Institute for Genomics and Evolutionary Medicine, Temple University, Philadelphia, PA, USA.,Department of Biology, Temple University, Philadelphia, PA, USA.,Center for Excellence in Genome Medicine and Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
75
|
Zucker MR, Abruzzo LV, Herling CD, Barron LL, Keating MJ, Abrams ZB, Heerema N, Coombes KR. Inferring clonal heterogeneity in cancer using SNP arrays and whole genome sequencing. Bioinformatics 2019; 35:2924-2931. [PMID: 30689715 PMCID: PMC6736450 DOI: 10.1093/bioinformatics/btz057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 01/18/2023] Open
Abstract
MOTIVATION Clonal heterogeneity is common in many types of cancer, including chronic lymphocytic leukemia (CLL). Previous research suggests that the presence of multiple distinct cancer clones is associated with clinical outcome. Detection of clonal heterogeneity from high throughput data, such as sequencing or single nucleotide polymorphism (SNP) array data, is important for gaining a better understanding of cancer and may improve prediction of clinical outcome or response to treatment. Here, we present a new method, CloneSeeker, for inferring clinical heterogeneity from sequencing data, SNP array data, or both. RESULTS We generated simulated SNP array and sequencing data and applied CloneSeeker along with two other methods. We demonstrate that CloneSeeker is more accurate than existing algorithms at determining the number of clones, distribution of cancer cells among clones, and mutation and/or copy numbers belonging to each clone. Next, we applied CloneSeeker to SNP array data from samples of 258 previously untreated CLL patients to gain a better understanding of the characteristics of CLL tumors and to elucidate the relationship between clonal heterogeneity and clinical outcome. We found that a significant majority of CLL patients appear to have multiple clones distinguished by copy number alterations alone. We also found that the presence of multiple clones corresponded with significantly worse survival among CLL patients. These findings may prove useful for improving the accuracy of prognosis and design of treatment strategies. AVAILABILITY AND IMPLEMENTATION Code available on R-Forge: https://r-forge.r-project.org/projects/CloneSeeker/. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Mark R Zucker
- Department of Biomedical Informatics, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Lynne V Abruzzo
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Carmen D Herling
- Department I of Internal Medicine, CIO Köln-Bonn, and CECAD, University of Cologne, Cologne, Germany
| | - Lynn L Barron
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Texas, MD, USA
| | - Michael J Keating
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Texas, MD, USA
| | - Zachary B Abrams
- Department of Biomedical Informatics, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Nyla Heerema
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Kevin R Coombes
- Department of Biomedical Informatics, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
76
|
Allogeneic transplantation in multiple myeloma: long-term follow-up and cytogenetic subgroup analysis. Leukemia 2019; 33:2710-2719. [DOI: 10.1038/s41375-019-0537-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 05/29/2019] [Indexed: 12/16/2022]
|
77
|
Vlummens P, De Veirman K, Menu E, De Bruyne E, Offner F, Vanderkerken K, Maes K. The Use of Murine Models for Studying Mechanistic Insights of Genomic Instability in Multiple Myeloma. Front Genet 2019; 10:740. [PMID: 31475039 PMCID: PMC6704229 DOI: 10.3389/fgene.2019.00740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by the accumulation of clonal plasma cells in the bone marrow. In normal plasma cell development, cells undergo programmed DNA breaks and translocations, a process necessary for generation of a wide repertoire of antigen-specific antibodies. This process also makes them vulnerable for the acquisition of chromosomal defects. Well-known examples of these aberrations, already seen at time of MM diagnosis, are hyperdiploidy or the translocations involving the immunoglobulin heavy chain. Over the recent years, however, novel aspects concerning genomic instability and its role in tumor development, disease progression and nascence of refractory disease were identified. As such, genomic instability is becoming a very relevant research topic with the potential identification of novel disease pathways. In this review, we aim to describe recent studies involving murine MM models focusing on the deregulation of processes implicated in genomic instability and their clinical impact. More specifically, we will discuss chromosomal instability, DNA damage and repair responses, development of drug resistance, and recent insights into the study of clonal hierarchy using different murine MM models. Lastly, we will discuss the importance and the use of murine MM models in the pre-clinical evaluation of promising novel therapeutic agents.
Collapse
Affiliation(s)
- Philip Vlummens
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Clinical Hematology, Ghent University Hospital, Gent, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fritz Offner
- Department of Clinical Hematology, Ghent University Hospital, Gent, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
78
|
Hierarchy of mono- and biallelic TP53 alterations in multiple myeloma cell fitness. Blood 2019; 134:836-840. [PMID: 31340981 DOI: 10.1182/blood.2019000080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
79
|
High-Risk Multiple Myeloma: Integrated Clinical and Omics Approach Dissects the Neoplastic Clone and the Tumor Microenvironment. J Clin Med 2019; 8:jcm8070997. [PMID: 31323969 PMCID: PMC6678140 DOI: 10.3390/jcm8070997] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a genetically heterogeneous disease that includes a subgroup of 10–15% of patients facing dismal survival despite the most intensive treatment. Despite improvements in biological knowledge, MM is still an incurable neoplasia, and therapeutic options able to overcome the relapsing/refractory behavior represent an unmet clinical need. The aim of this review is to provide an integrated clinical and biological overview of high-risk MM, discussing novel therapeutic perspectives, targeting the neoplastic clone and its microenvironment. The dissection of the molecular determinants of the aggressive phenotypes and drug-resistance can foster a better tailored clinical management of the high-risk profile and therapy-refractoriness. Among the current clinical difficulties in MM, patients’ management by manipulating the tumor niche represents a major challenge. The angiogenesis and the stromal infiltrate constitute pivotal mechanisms of a mutual collaboration between MM and the non-tumoral counterpart. Immuno-modulatory and anti-angiogenic therapy hold great efficacy, but variable and unpredictable responses in high-risk MM. The comprehensive understanding of the genetic heterogeneity and MM high-risk ecosystem enforce a systematic bench-to-bedside approach. Here, we provide a broad outlook of novel druggable targets. We also summarize the existing multi-omics-based risk profiling tools, in order to better select candidates for dual immune/vasculogenesis targeting.
Collapse
|
80
|
Abstract
To a large extent, cancer conforms to evolutionary rules defined by the rates at which clones mutate, adapt and grow. Next-generation sequencing has provided a snapshot of the genetic landscape of most cancer types, and cancer genomics approaches are driving new insights into cancer evolutionary patterns in time and space. In contrast to species evolution, cancer is a particular case owing to the vast size of tumour cell populations, chromosomal instability and its potential for phenotypic plasticity. Nevertheless, an evolutionary framework is a powerful aid to understand cancer progression and therapy failure. Indeed, such a framework could be applied to predict individual tumour behaviour and support treatment strategies.
Collapse
Affiliation(s)
- Samra Turajlic
- Cancer Evolution and Genome Instability Laboratory, Francis Crick Institute, London, UK
- Skin and Renal Units, The Royal Marsden NHS Foundation Trust, London, UK
| | - Andrea Sottoriva
- Evolutionary Genomics and Modelling Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Trevor Graham
- Tumour Biology, Evolution and Cancer Laboratory, Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence London, University College London Cancer Institute, London, UK.
- Department of Medical Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
81
|
Jones JR, Weinhold N, Ashby C, Walker BA, Wardell C, Pawlyn C, Rasche L, Melchor L, Cairns DA, Gregory WM, Johnson D, Begum DB, Ellis S, Sherborne AL, Cook G, Kaiser MF, Drayson MT, Owen RG, Jackson GH, Davies FE, Greaves M, Morgan GJ. Clonal evolution in myeloma: the impact of maintenance lenalidomide and depth of response on the genetics and sub-clonal structure of relapsed disease in uniformly treated newly diagnosed patients. Haematologica 2019; 104:1440-1450. [PMID: 30733268 PMCID: PMC6601103 DOI: 10.3324/haematol.2018.202200] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/30/2019] [Indexed: 12/27/2022] Open
Abstract
The emergence of treatment resistant sub-clones is a key feature of relapse in multiple myeloma. Therapeutic attempts to extend remission and prevent relapse include maximizing response and the use of maintenance therapy. We used whole exome sequencing to study the genetics of paired samples taken at presentation and at relapse from 56 newly diagnosed patients, following induction therapy, randomized to receive either lenalidomide maintenance or observation as part of the Myeloma XI trial. Patients included were considered high risk, relapsing within 30 months of maintenance randomization. Patients achieving a complete response had predominantly branching evolutionary patterns leading to relapse, characterized by a greater mutational burden, an altered mutational profile, bi-allelic inactivation of tumor suppressor genes, and acquired structural aberrations. Conversely, in patients achieving a partial response, the evolutionary features were predominantly stable with a similar mutational and structural profile seen at both time points. There were no significant differences between patients relapsing after lenalidomide maintenance versus observation. This study shows that the depth of response is a key determinant of the evolutionary patterns seen at relapse. This trial is registered at clinicaltrials.gov identifier: 01554852.
Collapse
Affiliation(s)
- John R Jones
- Department of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Cody Ashby
- 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
| | - Chris Wardell
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charlotte Pawlyn
- Department of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Leo Rasche
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - David A Cairns
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, UK
| | - Walter M Gregory
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, UK
| | | | - Dil B Begum
- The Institute of Cancer Research, London, UK
| | - Sidra Ellis
- The Institute of Cancer Research, London, UK
| | - Amy L Sherborne
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gordon Cook
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Martin F Kaiser
- Department of Haematology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Institute of Cancer Research, London, UK
| | - Mark T Drayson
- Clinical Immunology, School of Immunity and Infection, University of Birmingham, UK
| | - Roger G Owen
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Graham H Jackson
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - Faith E Davies
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mel Greaves
- The Institute of Cancer Research, London, UK
| | - Gareth J Morgan
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
82
|
Barwick BG, Neri P, Bahlis NJ, Nooka AK, Dhodapkar MV, Jaye DL, Hofmeister CC, Kaufman JL, Gupta VA, Auclair D, Keats JJ, Lonial S, Vertino PM, Boise LH. Multiple myeloma immunoglobulin lambda translocations portend poor prognosis. Nat Commun 2019; 10:1911. [PMID: 31015454 PMCID: PMC6478743 DOI: 10.1038/s41467-019-09555-6] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
Multiple myeloma is a malignancy of antibody-secreting plasma cells. Most patients benefit from current therapies, however, 20% of patients relapse or die within two years and are deemed high risk. Here we analyze structural variants from 795 newly-diagnosed patients as part of the CoMMpass study. We report translocations involving the immunoglobulin lambda (IgL) locus are present in 10% of patients, and indicative of poor prognosis. This is particularly true for IgL-MYC translocations, which coincide with focal amplifications of enhancers at both loci. Importantly, 78% of IgL-MYC translocations co-occur with hyperdiploid disease, a marker of standard risk, suggesting that IgL-MYC-translocated myeloma is being misclassified. Patients with IgL-translocations fail to benefit from IMiDs, which target IKZF1, a transcription factor that binds the IgL enhancer at some of the highest levels in the myeloma epigenome. These data implicate IgL translocation as a driver of poor prognosis which may be due to IMiD resistance. Multiple myeloma is frequently characterised by translocation of genes next to the immunoglobulin heavy chain locus. In this study, the authors sequence a large cohort of high risk myeloma samples and find translocations of cMyc to the immunoglobulin heavy chain locus and this is associated with poor prognosis.
Collapse
Affiliation(s)
- Benjamin G Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Paola Neri
- Charbonneau Cancer Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Nizar J Bahlis
- Charbonneau Cancer Research Institute, University of Calgary, 3330 Hospital Drive, Calgary, AB, T2N 4N1, Canada
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - David L Jaye
- Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Craig C Hofmeister
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Vikas A Gupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Daniel Auclair
- Multiple Myeloma Research Foundation, 383 Main Avenue, 5th Floor, Norwalk, CT, 06851, USA
| | - Jonathan J Keats
- Translational Genomics Research Institute, 445 North Fifth Street, Phoenix, AZ, 85004, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA.,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Paula M Vertino
- Department of Radiation Oncology, Emory University School of Medicine, 1701 Uppergate Drive, Atlanta, GA, 30322, USA. .,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA. .,Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA.
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365 Clifton Rd. NE, Atlanta, GA, 30322, USA. .,Winship Cancer Institute, Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA.
| |
Collapse
|
83
|
Hodgkinson A, Le Cam L, Trucu D, Radulescu O. Spatio-Genetic and phenotypic modelling elucidates resistance and re-sensitisation to treatment in heterogeneous melanoma. J Theor Biol 2019; 466:84-105. [PMID: 30503930 DOI: 10.1016/j.jtbi.2018.11.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 11/06/2018] [Accepted: 11/29/2018] [Indexed: 12/11/2022]
|
84
|
Lübke J, Naumann N, Kluger S, Schwaab J, Metzgeroth G, Evans E, Gardino AK, Lengauer C, Hofmann WK, Fabarius A, Cross NCP, Reiter A, Jawhar M. Inhibitory effects of midostaurin and avapritinib on myeloid progenitors derived from patients with KIT D816V positive advanced systemic mastocytosis. Leukemia 2019; 33:1195-1205. [PMID: 30911112 PMCID: PMC6756065 DOI: 10.1038/s41375-019-0450-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 02/03/2023]
Abstract
Advanced systemic mastocytosis (advSM) is characterized by the presence of an acquired KIT D816V mutation in >90% of patients. In the majority of patients, KIT D816V is not only detected in mast cells but also in other hematopoietic lineages. We sought to investigate the effects of the KIT-inhibitors midostaurin and avapritinib on single-cell-derived myeloid progenitor cells using granulocyte-macrophage colony-forming-units of patients with KIT D816V positive advSM. Colonies obtained prior to treatment were incubated in vitro with midostaurin (n = 10) or avapritinib (n = 11) and showed a marked reduction (≥50%) of KIT D816V positive colonies in 3/10 (30%) and 7/11 (64%) patient samples, respectively. Three of those 7 (43%) avapritinib responders were resistant to midostaurin in both, in vitro and in vivo. Colonies from four patients with high-risk molecular profile and aggressive clinical course were resistant to both drugs. The in vitro activity of midostaurin strongly correlated with clinical and molecular responses, e.g., relative reduction of KIT D816V allele burden and the proportion of KIT D816V positive colonies obtained after six months midostaurin-treatment in vivo. We conclude that the colony inhibition assay provides useful information for prediction of responses on midostaurin and that avapritinib has a superior in vitro activity compared to midostaurin.
Collapse
Affiliation(s)
- Johannes Lübke
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Nicole Naumann
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Sebastian Kluger
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Juliana Schwaab
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Georgia Metzgeroth
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Erica Evans
- Blueprint Medicines Corporation, Cambridge, MA, USA
| | | | | | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Alice Fabarius
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| | - Nicholas C P Cross
- Wessex Regional Genetics Laboratory, Salisbury NHS Foundation Trust, Salisbury, UK.,Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andreas Reiter
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany.
| | - Mohamad Jawhar
- Department of Hematology and Oncology, University Medical Centre Mannheim, Mannheim, Germany
| |
Collapse
|
85
|
Rasche L, Kortüm KM, Raab MS, Weinhold N. The Impact of Tumor Heterogeneity on Diagnostics and Novel Therapeutic Strategies in Multiple Myeloma. Int J Mol Sci 2019; 20:ijms20051248. [PMID: 30871078 PMCID: PMC6429294 DOI: 10.3390/ijms20051248] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/31/2022] Open
Abstract
Myeloma is characterized by extensive inter-patient genomic heterogeneity due to multiple different initiating events. A recent multi-region sequencing study demonstrated spatial differences, with progression events, such as TP53 mutations, frequently being restricted to focal lesions. In this review article, we describe the clinical impact of these two types of tumor heterogeneity. Target mutations are often dominant at one site but absent at other sites, which poses a significant challenge to personalized therapy in myeloma. The same holds true for high-risk subclones, which can be locally restricted, and as such not detectable at the iliac crest, which is the usual sampling site. Imaging can improve current risk classifiers and monitoring of residual disease, but does not allow for deciphering the molecular characteristics of tumor clones. In the era of novel immunotherapies, the clinical impact of heterogeneity certainly needs to be re-defined. Yet, preliminary observations indicate an ongoing impact of spatial heterogeneity on the efficacy of monoclonal antibodies. In conclusion, we recommend combining molecular tests with imaging to improve risk prediction and monitoring of residual disease. Overcoming intra-tumor heterogeneity is the prerequisite for curing myeloma. Novel immunotherapies are promising but research addressing their impact on the spatial clonal architecture is highly warranted.
Collapse
Affiliation(s)
- Leo Rasche
- Department of Internal Medicine 2, University Hospital of Würzburg, 97080 Würzburg, Germany.
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - K Martin Kortüm
- Department of Internal Medicine 2, University Hospital of Würzburg, 97080 Würzburg, Germany.
| | - Marc S Raab
- Department of Internal Medicine V, University Hospital of Heidelberg, 69120 Heidelberg, Germany.
| | - Niels Weinhold
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Internal Medicine V, University Hospital of Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
86
|
Dzobo K, Senthebane DA, Thomford NE, Rowe A, Dandara C, Parker MI. Not Everyone Fits the Mold: Intratumor and Intertumor Heterogeneity and Innovative Cancer Drug Design and Development. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:17-34. [PMID: 29356626 DOI: 10.1089/omi.2017.0174] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Disruptive innovations in medicine are game-changing in nature and bring about radical shifts in the way we understand human diseases, their treatment, and/or prevention. Yet, disruptive innovations in cancer drug design and development are still limited. Therapies that cure all cancer patients are in short supply or do not exist at all. Chief among the causes of this predicament is drug resistance, a mechanism that is much more dynamic than previously understood. Drug resistance has limited the initial success experienced with biomarker-guided targeted therapies as well. A major contributor to drug resistance is intratumor heterogeneity. For example, within solid tumors, there are distinct subclones of cancer cells, presenting profound complexity to cancer treatment. Well-known contributors to intratumor heterogeneity are genomic instability, the microenvironment, cellular genotype, cell plasticity, and stochastic processes. This expert review explains that for oncology drug design and development to be more innovative, we need to take into account intratumor heterogeneity. Initially thought to be the preserve of cancer cells, recent evidence points to the highly heterogeneous nature and diverse locations of stromal cells, such as cancer-associated fibroblasts (CAFs) and cancer-associated macrophages (CAMs). Distinct subpopulations of CAFs and CAMs are now known to be located immediately adjacent and distant from cancer cells, with different subpopulations exerting different effects on cancer cells. Disruptive innovation and precision medicine in clinical oncology do not have to be a distant reality, but can potentially be achieved by targeting these spatially separated and exclusive cancer cell subclones and CAF subtypes. Finally, we emphasize that disruptive innovations in drug discovery and development will likely come from drugs whose effect is not necessarily tumor shrinkage.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Dimakatso Alice Senthebane
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Nicholas Ekow Thomford
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Arielle Rowe
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa
| | - Collet Dandara
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - M Iqbal Parker
- 2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
87
|
Harding T, Baughn L, Kumar S, Van Ness B. The future of myeloma precision medicine: integrating the compendium of known drug resistance mechanisms with emerging tumor profiling technologies. Leukemia 2019; 33:863-883. [PMID: 30683909 DOI: 10.1038/s41375-018-0362-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/25/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy that is considered mostly incurable in large part due to the inability of standard of care therapies to overcome refractory disease and inevitable drug-resistant relapse. The post-genomic era has been a productive period of discovery where modern sequencing methods have been applied to large MM patient cohorts to modernize our current perception of myeloma pathobiology and establish an appreciation for the vast heterogeneity that exists between and within MM patients. Numerous pre-clinical studies conducted in the last two decades have unveiled a compendium of mechanisms by which malignant plasma cells can escape standard therapies, many of which have potentially quantifiable biomarkers. Exhaustive pre-clinical efforts have evaluated countless putative anti-MM therapeutic agents and many of these have begun to enter clinical trial evaluation. While the palette of available anti-MM therapies is continuing to expand it is also clear that malignant plasma cells still have mechanistic avenues by which they can evade even the most promising new therapies. It is therefore becoming increasingly clear that there is an outstanding need to develop and employ precision medicine strategies in MM management that harness emerging tumor profiling technologies to identify biomarkers that predict efficacy or resistance within an individual's sub-clonally heterogeneous tumor. In this review we present an updated overview of broad classes of therapeutic resistance mechanisms and describe selected examples of putative biomarkers. We also outline several emerging tumor profiling technologies that have the potential to accurately quantify biomarkers for therapeutic sensitivity and resistance at genomic, transcriptomic and proteomic levels. Finally, we comment on the future of implementation for precision medicine strategies in MM and the clear need for a paradigm shift in clinical trial design and disease management.
Collapse
Affiliation(s)
- Taylor Harding
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, USA
| | - Linda Baughn
- Department of Laboratory Medicine and Pathology, Division of Laboratory Genetics, Mayo Clinic, Rochester, MN, USA
| | - Shaji Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic Rochester, Rochester, USA
| | - Brian Van Ness
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
88
|
Jang JS, Li Y, Mitra AK, Bi L, Abyzov A, van Wijnen AJ, Baughn LB, Van Ness B, Rajkumar V, Kumar S, Jen J. Molecular signatures of multiple myeloma progression through single cell RNA-Seq. Blood Cancer J 2019; 9:2. [PMID: 30607001 PMCID: PMC6318319 DOI: 10.1038/s41408-018-0160-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/17/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022] Open
Abstract
We used single cell RNA-Seq to examine molecular heterogeneity in multiple myeloma (MM) in 597 CD138 positive cells from bone marrow aspirates of 15 patients at different stages of disease progression. 790 genes were selected by coefficient of variation (CV) method and organized cells into four groups (L1–L4) using unsupervised clustering. Plasma cells from each patient clustered into at least two groups based on gene expression signature. The L1 group contained cells from all MGUS patients having the lowest expression of genes involved in the oxidative phosphorylation, Myc targets, and mTORC1 signaling pathways (p < 1.2 × 10−14). In contrast, the expression level of these pathway genes increased progressively and were the highest in L4 group containing only cells from MM patients with t(4;14) translocations. A 44 genes signature of consistently overexpressed genes among the four groups was associated with poorer overall survival in MM patients (APEX trial, p < 0.0001; HR, 1.83; 95% CI, 1.33–2.52), particularly those treated with bortezomib (p < 0.0001; HR, 2.00; 95% CI, 1.39–2.89). Our study, using single cell RNA-Seq, identified the most significantly affected molecular pathways during MM progression and provided a novel signature predictive of patient prognosis and treatment stratification.
Collapse
Affiliation(s)
- Jin Sung Jang
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ying Li
- Division of Bioinformatics and Biostatistics, Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Lintao Bi
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Alexej Abyzov
- Division of Bioinformatics and Biostatistics, Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | | | - Linda B Baughn
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Brian Van Ness
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, USA
| | - Vincent Rajkumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shaji Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Jin Jen
- Genome Analysis Core, Medical Genome Facility, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA. .,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
89
|
Vikova V, Jourdan M, Robert N, Requirand G, Boireau S, Bruyer A, Vincent L, Cartron G, Klein B, Elemento O, Kassambara A, Moreaux J. Comprehensive characterization of the mutational landscape in multiple myeloma cell lines reveals potential drivers and pathways associated with tumor progression and drug resistance. Theranostics 2019; 9:540-553. [PMID: 30809292 PMCID: PMC6376179 DOI: 10.7150/thno.28374] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022] Open
Abstract
Human multiple myeloma tumor cell lines (HMCLs) have been a cornerstone of research in multiple myeloma (MM) and have helped to shape our understanding of molecular processes that drive tumor progression. A comprehensive characterization of genomic mutations in HMCLs will provide a basis for choosing relevant cell line models to study a particular aspect of myeloma biology, or to screen for an antagonist of certain cancer pathways. Methods: We performed whole exome sequencing on a large cohort of 30 HMCLs, representative of a large molecular heterogeneity of MM, and 8 control samples (epstein-barr virus (EBV)-immortalized B-cells obtained from 8 different patients). We evaluated the sensitivity of HMCLs to ten drugs. Results: We identified a high confidence list of 236 protein-coding genes with mutations affecting the structure of the encoded protein. Among the most frequently mutated genes, there were known MM drivers, such as TP53, KRAS, NRAS, ATM and FAM46C, as well as novel mutated genes, including CNOT3, KMT2D, MSH3 and PMS1. We next generated a comprehensive map of altered key pathways in HMCLs. These include cell growth pathways (MAPK, JAK-STAT, PI(3)K-AKT and TP53 / cell cycle pathway), DNA repair pathway and chromatin modifiers. Importantly, our analysis highlighted a significant association between the mutation of several genes and the response to conventional drugs used in MM as well as targeted inhibitors. Conclusion: Taken together, this first comprehensive exome-wide analysis of the mutational landscape in HMCLs provides unique resources for further studies and identifies novel genes potentially associated with MM pathophysiology, some of which may be targets for future therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Nicolas Robert
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | - Guilhem Requirand
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | - Stéphanie Boireau
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | | | - Laure Vincent
- CHU Montpellier, Department of Clinical Hematology, Montpellier, France
| | - Guillaume Cartron
- Univ Montpellier, UFR de Médecine, Montpellier, France
- CHU Montpellier, Department of Clinical Hematology, Montpellier, France
- Univ Montpellier, UMR CNRS 5235, Montpellier, France
| | - Bernard Klein
- IGH, CNRS, Univ Montpellier, France
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
- Univ Montpellier, UFR de Médecine, Montpellier, France
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, 1305 York Avenue, New York, NY 10021, USA
| | - Alboukadel Kassambara
- IGH, CNRS, Univ Montpellier, France
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | - Jérôme Moreaux
- IGH, CNRS, Univ Montpellier, France
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
- Univ Montpellier, UFR de Médecine, Montpellier, France
| |
Collapse
|
90
|
Single-cell analysis identifies CRLF2 rearrangements as both early and late events in Down syndrome and non-Down syndrome acute lymphoblastic leukaemia. Leukemia 2018; 33:893-904. [PMID: 30487598 PMCID: PMC6398588 DOI: 10.1038/s41375-018-0297-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/19/2018] [Accepted: 09/24/2018] [Indexed: 12/23/2022]
Abstract
Deregulated expression of the type I cytokine receptor, CRLF2, is observed in 5-15% of precursor B-cell acute lymphoblastic leukaemia (B-ALL). We have previously reported the genomic landscape of patients with CRLF2 rearrangements (CRLF2-r) using both whole genome and exome sequencing, which identified a number of potential clonal and sub-clonal genomic alterations. In this study, we aimed to assess when the CRLF2-r; IGH-CRLF2 or P2RY8-CRLF2, arose during the evolution of both Down syndrome-ALL (DS-ALL) and non-DS-ALL. Using fluorescence in situ hybridisation, we were able to track up to four structural variants in single cells from 47 CRLF2-r B-ALL patients, which in association with our multiplex single cell analysis of a further four patients, permitted simultaneous tracking of copy number alterations, structural and single nucleotide variants within individual cells. We observed CRLF2-r arising as both early and late events in DS and non-DS-ALL patients. Parallel evolution of discrete clones was observed in the development of CRLF2-r B-ALL, either involving the CRLF2-r or one of the other tracked abnormalities. In depth single cell analysis identified both linear and branching evolution with early clones harbouring a multitude of abnormalities, including the CRLF2-r in DS-ALL patients.
Collapse
|
91
|
Xu J, Wang Q, Xu H, Gu C, Jiang L, Wang J, Wang D, Xu B, Mao X, Wang J, Wang Z, Xiao Y, Zhang Y, Li C, Zhou J. Anti-BCMA CAR-T cells for treatment of plasma cell dyscrasia: case report on POEMS syndrome and multiple myeloma. J Hematol Oncol 2018; 11:128. [PMID: 30348186 PMCID: PMC6198365 DOI: 10.1186/s13045-018-0672-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/14/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND POEMS (polyneuropathy, organomegaly, endocrinopathy, monoclonal gammopathy, and skin changes) syndrome still has no standard treatment. On the basis that both POEMS syndrome and myeloma have an underlying plasma cell dyscrasia, anti-myeloma therapy can be expected to be useful for POEMS syndrome. Chimeric antigen receptor T (CAR-T) cells targeting B cell maturation antigen (BCMA) has been used in the treatment of relapsed and refractory multiple myeloma (RRMM). No POEMS syndrome cases treated with anti-BCMA CAR-T cells have been reported. CASE PRESENTATION Here, we, for the first time, report a POEMS syndrome case treated with anti-BCMA CAR-T cells. A 49-year-old female with incapacitating POEMS syndrome that progressed on lenalidomide treatment was enrolled in a phase I study involving anti-BCMA CAR-T cells (ChiCTR-OPC-16009113). Another patient with RRMM who had undergone six prior lines treatments was also enrolled in the study. They received infusions of anti-BCMA CAR-T cells. Both patients achieved a stringent complete response. Complete remission persisted in the patient with POEMS syndrome and lasted for 7.6 months before a relapse in RRMM patient. Both patients had toxicity consistent with the grade 1 cytokine release syndrome. CONCLUSIONS This is the first report of treatment by anti-BCMA CAR-T cells in POEMS syndrome. Our findings demonstrate the anti-BCMA CAR-T cell treatment may be a feasible therapeutic option for patients with POEMS syndrome and RRMM who do not respond well to traditional therapies. TRIAL REGISTRATION ChiCTR-OPC, ChiCTR-OPC-16009113 . Registered 29 August 2016.
Collapse
Affiliation(s)
- Jinhuan Xu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Qiuxiang Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Hao Xu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Chaojiang Gu
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China
| | - Lijun Jiang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Jue Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Di Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Bin Xu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Xia Mao
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Jin Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Zhiqiong Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, 1095 Jie-Fang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
92
|
Adamik J, Galson DL, Roodman GD. Osteoblast suppression in multiple myeloma bone disease. J Bone Oncol 2018; 13:62-70. [PMID: 30591859 PMCID: PMC6303385 DOI: 10.1016/j.jbo.2018.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022] Open
Abstract
Multiple myeloma (MM) is the most frequent cancer to involve the skeleton with patients developing osteolytic bone lesions due to hyperactivation of osteoclasts and suppression of BMSCs differentiation into functional osteoblasts. Although new therapies for MM have greatly improved survival, MM remains incurable for most patients. Despite the major advances in current anti-MM and anti-resorptive treatments that can significantly improve osteolytic bone lysis, many bone lesions can persist even after therapeutic remission of active disease. Bone marrow mesenchymal stem cells (BMSCs) from MM patients are phenotypically distinct from their healthy counterparts and the mechanisms associated with the long-term osteogenic suppression are largely unknown. In this review we will highlight recent results of transcriptomic profiling studies that provide new insights into the establishment and maintenance of the persistent pathological alterations in MM-BMSCs that occur in MM. We will we discuss the role of genomic instabilities and senescence in propagating the chronically suppressed state and pro-inflammatory phenotype associated with MM-BMSCs. Lastly we describe the role of epigenetic-based mechanisms in regulating osteogenic gene expression to establish and maintain the pro-longed suppression of MM-BMSC differentiation into functional OBs.
Collapse
Affiliation(s)
- Juraj Adamik
- Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine University of Pittsburgh, Pittsburgh, PA, USA
| | - Deborah L Galson
- Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, The McGowan Institute for Regenerative Medicine University of Pittsburgh, Pittsburgh, PA, USA
| | - G David Roodman
- Department of Medicine, Division of Hematology-Oncology, Indiana University, Indianapolis, IN, USA.,Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
93
|
Caers J, Garderet L, Kortüm KM, O'Dwyer ME, van de Donk NWCJ, Binder M, Dold SM, Gay F, Corre J, Beguin Y, Ludwig H, Larocca A, Driessen C, Dimopoulos MA, Boccadoro M, Gramatzki M, Zweegman S, Einsele H, Cavo M, Goldschmidt H, Sonneveld P, Delforge M, Auner HW, Terpos E, Engelhardt M. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: what to use and when. Haematologica 2018; 103:1772-1784. [PMID: 30171031 PMCID: PMC6278986 DOI: 10.3324/haematol.2018.189159] [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: 01/21/2018] [Accepted: 08/27/2018] [Indexed: 01/04/2023] Open
Abstract
The diagnosis of multiple myeloma can be challenging, even for experienced physicians, and requires close collaboration between numerous disciplines (orthopedics, radiology, nuclear medicine, radiation therapy, hematology and oncology) before the final diagnosis of myeloma is made. The definition of multiple myeloma is based on the presence of clinical, biochemical, histopathological, and radiological markers of disease. Specific tests are needed both at presentation and during follow-up in order to reach the correct diagnosis and characterize the disease precisely. These tests can also serve prognostic purposes and are useful for follow-up of myeloma patients. Molecular analyses remain pivotal for defining high-risk myeloma and are used in updated patient stratifications, while minimal residual disease assessment via flow cytometry, molecular techniques and radiological approaches provides additional prognostic information on patients' long-term outcome. This pivotal information will guide our future treatment decisions in forthcoming clinical trials. The European Myeloma Network group updated their guidelines on different diagnostic recommendations, which should be of value to enable appropriate use of the recommendations both at diagnosis and during follow-up.
Collapse
Affiliation(s)
- Jo Caers
- Department of Hematology, University Hospital of Liege, Belgium .,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | | | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michael E O'Dwyer
- Department of Hematology, National University of Ireland Galway, Ireland
| | | | - Mascha Binder
- Department of Internal Medicine II, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sandra Maria Dold
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Francesca Gay
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Jill Corre
- Unit for Genomics in Myeloma, Institut Universitaire du Cancer - Oncopole, Toulouse, France
| | - Yves Beguin
- Department of Hematology, University Hospital of Liege, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Belgium
| | - Heinz Ludwig
- Wilhelminen Cancer Research Institute, Vienna, Austria
| | - Alessandra Larocca
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Christoph Driessen
- Department of Oncology and Hematology, Cantonal Hospital St. Gallen, Switzerland
| | | | - Mario Boccadoro
- Department of Hematology-Oncology, University Hospital Città della Salute e della Scienza, Torino, Italy
| | - Martin Gramatzki
- Division of Stem Cell Transplantation and Immunotherapy, University of Kiel, Germany
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Wuerzburg, Germany
| | - Michele Cavo
- Seragnoli 'Institute of Hematology, Bologna University School of Medicine, Italy
| | - Hartmut Goldschmidt
- Department of Hematology, Rheumatology and Oncology, University Hospital Heidelberg, Germany.,National Center for Tumor Diseases, Heidelberg Medical University, Germany
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Michel Delforge
- Department of Hematology, University Hospital Leuven, Belgium
| | - Holger W Auner
- Centre for Haematology, Hammersmith Hospital, Imperial College London, UK
| | - Evangelos Terpos
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
94
|
Brudno JN, Maric I, Hartman SD, Rose JJ, Wang M, Lam N, Stetler-Stevenson M, Salem D, Yuan C, Pavletic S, Kanakry JA, Ali SA, Mikkilineni L, Feldman SA, Stroncek DF, Hansen BG, Lawrence J, Patel R, Hakim F, Gress RE, Kochenderfer JN. T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J Clin Oncol 2018; 36:2267-2280. [PMID: 29812997 PMCID: PMC6067798 DOI: 10.1200/jco.2018.77.8084] [Citation(s) in RCA: 600] [Impact Index Per Article: 85.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Purpose Therapies with novel mechanisms of action are needed for multiple myeloma (MM). T cells can be genetically modified to express chimeric antigen receptors (CARs), which are artificial proteins that target T cells to antigens. B-cell maturation antigen (BCMA) is expressed by normal and malignant plasma cells but not normal essential cells. We conducted the first-in-humans clinical trial, to our knowledge, of T cells expressing a CAR targeting BCMA (CAR-BCMA). Patients and Methods Sixteen patients received 9 × 106 CAR-BCMA T cells/kg at the highest dose level of the trial; we are reporting results of these 16 patients. The patients had a median of 9.5 prior lines of MM therapy. Sixty-three percent of patients had MM refractory to the last treatment regimen before protocol enrollment. T cells were transduced with a γ-retroviral vector encoding CAR-BCMA. Patients received CAR-BCMA T cells after a conditioning chemotherapy regimen of cyclophosphamide and fludarabine. Results The overall response rate was 81%, with 63% very good partial response or complete response. Median event-free survival was 31 weeks. Responses included eradication of extensive bone marrow myeloma and resolution of soft-tissue plasmacytomas. All 11 patients who obtained an anti-MM response of partial response or better and had MM evaluable for minimal residual disease obtained bone marrow minimal residual disease-negative status. High peak blood CAR+ cell levels were associated with anti-MM responses. Cytokine-release syndrome toxicities were severe in some cases but were reversible. Blood CAR-BCMA T cells were predominantly highly differentiated CD8+ T cells 6 to 9 days after infusion. BCMA antigen loss from MM was observed. Conclusion CAR-BCMA T cells had substantial activity against heavily treated relapsed/refractory MM. Our results should encourage additional development of CAR T-cell therapies for MM.
Collapse
Affiliation(s)
- Jennifer N. Brudno
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Irina Maric
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Steven D. Hartman
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Jeremy J. Rose
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Michael Wang
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Norris Lam
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Maryalice Stetler-Stevenson
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Dalia Salem
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Constance Yuan
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Steven Pavletic
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Jennifer A. Kanakry
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Syed Abbas Ali
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Lekha Mikkilineni
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Steven A. Feldman
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - David F. Stroncek
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Brenna G. Hansen
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Judith Lawrence
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Rashmika Patel
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Frances Hakim
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - Ronald E. Gress
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| | - James N. Kochenderfer
- Jennifer N. Brudno, Irina Maric, Steven D. Hartman, Jeremy J. Rose, Norris Lam, Maryalice Stetler-Stevenson, Dalia Salem, Constance Yuan, Steven Pavletic, Jennifer A. Kanakry, Lekha Mikkilineni, Steven A. Feldman, David F. Stroncek, Brenna G. Hansen, Rashmika Patel, Frances Hakim, Ronald E. Gress, and James N. Kochenderfer, National Institutes of Health, Bethesda; Syed Abbas Ali, Johns Hopkins University Sidney Kimmel Comprehensive Cancer Center, Baltimore; Judith Lawrence, Leidos Biomedical Research, Frederick, MD; Michael Wang, University of Texas MD Anderson Cancer Center, Houston, TX; and Dalia Salem, Mansoura University, Mansoura, Egypt
| |
Collapse
|
95
|
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
|
96
|
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
|
97
|
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
|
98
|
Linking transcriptional and genetic tumor heterogeneity through allele analysis of single-cell RNA-seq data. Genome Res 2018; 28:1217-1227. [PMID: 29898899 PMCID: PMC6071640 DOI: 10.1101/gr.228080.117] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 05/24/2018] [Indexed: 12/27/2022]
Abstract
Characterization of intratumoral heterogeneity is critical to cancer therapy, as the presence of phenotypically diverse cell populations commonly fuels relapse and resistance to treatment. Although genetic variation is a well-studied source of intratumoral heterogeneity, the functional impact of most genetic alterations remains unclear. Even less understood is the relative importance of other factors influencing heterogeneity, such as epigenetic state or tumor microenvironment. To investigate the relationship between genetic and transcriptional heterogeneity in a context of cancer progression, we devised a computational approach called HoneyBADGER to identify copy number variation and loss of heterozygosity in individual cells from single-cell RNA-sequencing data. By integrating allele and normalized expression information, HoneyBADGER is able to identify and infer the presence of subclone-specific alterations in individual cells and reconstruct the underlying subclonal architecture. By examining several tumor types, we show that HoneyBADGER is effective at identifying deletions, amplifications, and copy-neutral loss-of-heterozygosity events and is capable of robustly identifying subclonal focal alterations as small as 10 megabases. We further apply HoneyBADGER to analyze single cells from a progressive multiple myeloma patient to identify major genetic subclones that exhibit distinct transcriptional signatures relevant to cancer progression. Other prominent transcriptional subpopulations within these tumors did not line up with the genetic subclonal structure and were likely driven by alternative, nonclonal mechanisms. These results highlight the need for integrative analysis to understand the molecular and phenotypic heterogeneity in cancer.
Collapse
|
99
|
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
|
100
|
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
|