1
|
Hersby DS, Schejbel L, Breinholt MF, Høgdall E, Nørgaard P, Nielsen TH, Pedersen LM, Gang AO. Mutational heterogeneity in large B-cell lymphoma: insights from paired biopsies. Ann Hematol 2024:10.1007/s00277-024-06108-w. [PMID: 39644335 DOI: 10.1007/s00277-024-06108-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Large B-cell lymphoma (LBCL) exhibits striking clinical and molecular heterogeneity. New approaches have emerged to explore tumor heterogeneity and classify LBCL into biological categories. Consequently, the informational requirements from diagnostic samples to provide the necessary information have increased, but the adequacy of single-site biopsies to provide such information is largely unknown. Here we describe spatial and temporal intra-patient variations in the mutational landscape of paired biopsies. METHODS Paired biopsies from 30 patients with LBCL were obtained from spatially distinct sites at the time of primary diagnosis before treatment and/or at a subsequent relapse. The samples were sequenced using a custom designed 59-gene next generation sequencing (NGS) lymphoma panel. RESULTS Differences in detected mutations of pathogenic or likely pathogenic significance were frequent both when comparing paired diagnostic biopsies, 2/6 (33%), and when comparing paired biopsies at primary diagnosis and relapse, 8/16 (50%). Mutational heterogeneity tended to increase with longer time interval between biopsies. Analysis of paired diagnostic and relapse biopsies revealed that certain clones present at diagnosis disappeared, while new clones emerged at relapse. Notably, TP53 mutations were detected in six out of seven patients in an extranodal location. In two cases, TP53 mutation was only detected in the relapse biopsy. Several of the mutations identified in this study are used or under investigation as targets for cancer treatments. CONCLUSION Multi-site biopsies revealed spatial and temporal mutational heterogeneity in patients with LBCL. Our findings indicate that mutational differences between biopsy pairs can occur at all timepoints examined. This underscores the necessity of performing repeat biopsies with each relapse to capture the full spectrum of genetic aberrations.
Collapse
Affiliation(s)
| | - Lone Schejbel
- Department of Pathology, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Fredslund Breinholt
- Department of Pathology, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Estrid Høgdall
- Department of Pathology, Herlev Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter Nørgaard
- Department of Pathology, Hvidovre Hospitalet, Hvidovre, Denmark
| | - Torsten Holm Nielsen
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- Department of Hematology, Zealand Hospital, Roskilde, Denmark
- Danish Medicines Agency, Copenhagen, Denmark
| | - Lars Møller Pedersen
- Department of Hematology, Zealand Hospital, Roskilde, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Ortved Gang
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Mee MW, Faulkner S, Wood GA, Woods JP, Bienzle D, Coomber BL. Longitudinal Study of Transcriptomic Changes Occurring over Six Weeks of CHOP Treatment in Canine Lymphoma Identifies Prognostic Subtypes. Vet Sci 2024; 11:540. [PMID: 39591314 PMCID: PMC11599011 DOI: 10.3390/vetsci11110540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
The majority of canine lymphoma patients treated with the standard of care, the CHOP chemotherapy protocol, initially achieve remission but eventually relapse with a multi-drug-resistant phenotype. This study assesses gene expression profiles of canine lymphoma tumor cell populations using RNA-Seq data from 15 matched patient samples taken prior to treatment and again six weeks into treatment with CHOP. Two distinct clusters were present in the t-SNE dimensionality reduction of the gene expression profiles. There was a significant difference in progression-free survival (PFS) between the cluster groups, with a median of 43.5 days in a group of six patients and 185 days in another group of nine patients. Comparing the group with shorter PFS to the group with longer PFS, we identified 265 significantly enriched GO:BP terms in 3874 significantly up-regulated genes and 740 significantly enriched GO:BP terms in 3236 significantly down-regulated genes. Comparing the six-week timepoint against the initial timepoint, in the group with longer PFS, we identified 277 significantly enriched GO:BP terms in 413 significantly up-regulated genes and 222 significantly enriched GO:BP terms in 267 significantly down-regulated genes. In the group with shorter PFS, we only identified 27 significantly differentially expressed genes, for this comparison. We found DNA damage response genes to be enriched in the down-regulated genes in both comparisons. These results identify and characterize two transcriptionally distinct groups of canine lymphoma patients with significantly different responses to CHOP chemotherapy.
Collapse
Affiliation(s)
- Miles W. Mee
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Sydney Faulkner
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Geoffrey A. Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - J. Paul Woods
- Department of Clinical Studies and Mona Campbell Center for Animal Cancer, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Brenda L. Coomber
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
3
|
Fardoos R, Christensen C, Øbro NF, Overgaard UM, Als-Nielsen B, Madsen HO, Marquart HV. Flow Sorting, Whole Genome Amplification and Next-Generation Sequencing as Combined Tools to Study Heterogeneous Acute Lymphoblastic Leukemia. Diagnostics (Basel) 2023; 13:3306. [PMID: 37958202 PMCID: PMC10650172 DOI: 10.3390/diagnostics13213306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Next-generation sequencing (NGS) methods have been introduced for immunoglobulin (IG)/T-cell receptor (TR) gene rearrangement analysis in acute lymphoblastic leukemia (ALL) and lymphoma (LBL). These methods likely constitute faster and more sensitive approaches to analyze heterogenous cases of ALL/LBL, yet it is not known whether gene rearrangements constituting low percentages of the total sequence reads represent minor subpopulations of malignant cells or background IG/TR gene rearrangements in normal B-and T-cells. In a comparison of eight cases of B-cell precursor ALL (BCP-ALL) using both the EuroClonality NGS method and the IdentiClone multiplex-PCR/gene-scanning method, the NGS method identified between 29% and 139% more markers than the gene-scanning method, depending on whether the NGS data analysis used a threshold of 5% or 1%, respectively. As an alternative to using low thresholds, we show that IG/TR gene rearrangements in subpopulations of cancer cells can be discriminated from background IG/TR gene rearrangements in normal B-and T-cells through a combination of flow cytometry cell sorting and multiple displacement amplification (MDA)-based whole genome amplification (WGA) prior to the NGS. Using this approach to investigate the clonal evolution in a BCP-ALL patient with double relapse, clonal TR rearrangements were found in sorted leukemic cells at the time of second relapse that could be identified at the time of diagnosis, below 1% of the total sequence reads. These data emphasize that caution should be exerted when interpreting rare sequences in NGS experiments and show the advantage of employing the flow sorting of malignant cell populations in NGS clonality assessments.
Collapse
Affiliation(s)
- Rabiah Fardoos
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Claus Christensen
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Nina Friesgaard Øbro
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Ulrik Malthe Overgaard
- Department of Hematology, The University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Bodil Als-Nielsen
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Institute of Clinical Medicine, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Hans Ole Madsen
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Hanne Vibeke Marquart
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Institute of Clinical Medicine, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
4
|
Stuckey R, Luzardo Henríquez H, de la Nuez Melian H, Rivero Vera JC, Bilbao-Sieyro C, Gómez-Casares MT. Integration of molecular testing for the personalized management of patients with diffuse large B-cell lymphoma and follicular lymphoma. World J Clin Oncol 2023; 14:160-170. [PMID: 37124135 PMCID: PMC10134203 DOI: 10.5306/wjco.v14.i4.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/24/2023] [Accepted: 04/04/2023] [Indexed: 04/21/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) are the most common forms of aggressive and indolent lymphoma, respectively. The majority of patients are cured by standard R-CHOP immunochemotherapy, but 30%–40% of DLBCL and 20% of FL patients relapse or are refractory (R/R). DLBCL and FL are phenotypically and genetically hereterogenous B-cell neoplasms. To date, the diagnosis of DLBCL and FL has been based on morphology, immunophenotyping and cytogenetics. However, next-generation sequencing (NGS) is widening our understanding of the genetic basis of the B-cell lymphomas. In this review we will discuss how integrating the NGS-based characterization of somatic gene mutations with diagnostic or prognostic value in DLBCL and FL could help refine B-cell lymphoma classification as part of a multidisciplinary pathology work-up. We will also discuss how molecular testing can identify candidates for clinical trials with targeted therapies and help predict therapeutic outcome to currently available treatments, including chimeric antigen receptor T-cell, as well as explore the application of circulating cell-free DNA, a non-invasive method for patient monitoring. We conclude that molecular analyses can drive improvements in patient outcomes due to an increased understanding of the different pathogenic pathways affected by each DLBCL subtype and indolent FL vs R/R FL.
Collapse
Affiliation(s)
- Ruth Stuckey
- Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas 35019, Spain
| | - Hugo Luzardo Henríquez
- Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas 35019, Spain
| | | | - José Carlos Rivero Vera
- Department of Anatomical Pathology, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas 35019, Spain
| | - Cristina Bilbao-Sieyro
- Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrin, Las Palmas de Gran Canaria 35019, Las Palmas de Gran Canaria, Spain
- Department of Morphology, Universitario de Las Palmas de Gran Canaria, Las Palmas 35001, Spain
| | - María Teresa Gómez-Casares
- Department of Hematology, Hospital Universitario de Gran Canaria Dr. Negrin, Las Palmas de Gran Canaria 35019, Las Palmas de Gran Canaria, Spain
- Medical Science, Universitario de Las Palmas de Gran Canaria, Las Palmas 35001, Spain
| |
Collapse
|
5
|
Bewicke-Copley F, Korfi K, Araf S, Hodkinson B, Kumar E, Cummin T, Ashton-Key M, Barrans S, van Hoppe S, Burton C, Elshiekh M, Rule S, Crosbie N, Clear A, Calaminici M, Runge H, Hills RK, Scott DW, Rimsza LM, Menon G, Sha C, Davies JR, Nagano A, Davies A, Painter D, Smith A, Gribben J, Naresh KN, Westhead DR, Okosun J, Steele A, Hodson DJ, Balasubramanian S, Johnson P, Wang J, Fitzgibbon J. Longitudinal expression profiling identifies a poor risk subset of patients with ABC-type diffuse large B-cell lymphoma. Blood Adv 2023; 7:845-855. [PMID: 35947123 PMCID: PMC9986713 DOI: 10.1182/bloodadvances.2022007536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 07/05/2022] [Accepted: 07/25/2022] [Indexed: 11/20/2022] Open
Abstract
Despite the effectiveness of immuno-chemotherapy, 40% of patients with diffuse large B-cell lymphoma (DLBCL) experience relapse or refractory disease. Longitudinal studies have previously focused on the mutational landscape of relapse but fell short of providing a consistent relapse-specific genetic signature. In our study, we have focused attention on the changes in GEP accompanying DLBCL relapse using archival paired diagnostic/relapse specimens from 38 de novo patients with DLBCL. COO remained stable from diagnosis to relapse in 80% of patients, with only a single patient showing COO switching from activated B-cell-like (ABC) to germinal center B-cell-like (GCB). Analysis of the transcriptomic changes that occur following relapse suggest ABC and GCB relapses are mediated via different mechanisms. We developed a 30-gene discriminator for ABC-DLBCLs derived from relapse-associated genes that defined clinically distinct high- and low-risk subgroups in ABC-DLBCLs at diagnosis in datasets comprising both population-based and clinical trial cohorts. This signature also identified a population of <60-year-old patients with superior PFS and OS treated with ibrutinib-R-CHOP as part of the PHOENIX trial. Altogether this new signature adds to the existing toolkit of putative genetic predictors now available in DLBCL that can be readily assessed as part of prospective clinical trials.
Collapse
Affiliation(s)
- Findlay Bewicke-Copley
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Koorosh Korfi
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Shamzah Araf
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Brendan Hodkinson
- Oncology Translational Research, Janssen Research & Development, Spring House, PA
| | - Emil Kumar
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Thomas Cummin
- Cancer Research UK Centre, University of Southampton, Southampton, UK
| | - Margaret Ashton-Key
- Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sharon Barrans
- Haematological Malignancy Diagnostic Service, St. James’s Institute of Oncology, Leeds, UK
| | - Suzan van Hoppe
- Haematological Malignancy Diagnostic Service, St. James’s Institute of Oncology, Leeds, UK
| | - Cathy Burton
- Haematological Malignancy Diagnostic Service, St. James’s Institute of Oncology, Leeds, UK
| | - Mohamed Elshiekh
- Cellular & Molecular Pathology, Imperial College NHS Trust & Imperial College London, London, UK
| | - Simon Rule
- Department of Haematology, Derriford Hospital, University of Plymouth, Plymouth, UK
| | - Nicola Crosbie
- Department of Haematology, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Andrew Clear
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Maria Calaminici
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Hendrik Runge
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Robert K. Hills
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David W. Scott
- BC Cancer Centre for Lymphoid Cancer and Department of Medicine, University of British Columbia, Vancouver, BC Canada
| | - Lisa M. Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix AZ
| | - Geetha Menon
- Haemato-Oncology Diagnostic Service, Liverpool Clinical Laboratories, Liverpool, UK
| | - Chulin Sha
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - John R. Davies
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Ai Nagano
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Andrew Davies
- Cancer Research UK Centre, University of Southampton, Southampton, UK
| | - Daniel Painter
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York, UK
| | - Alexandra Smith
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York, UK
| | - John Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Kikkeri N. Naresh
- Cellular & Molecular Pathology, Imperial College NHS Trust & Imperial College London, London, UK
| | - David R. Westhead
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Andrew Steele
- Oncology Translational Research, Janssen Research & Development, San Diego, CA
| | - Daniel J. Hodson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Peter Johnson
- Cancer Research UK Centre, University of Southampton, Southampton, UK
| | - Jun Wang
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| | - Jude Fitzgibbon
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University, London, UK
| |
Collapse
|
6
|
Smith AL, Eiken AP, Skupa SA, Moore DY, Umeta LT, Smith LM, Lyden ER, D’Angelo CR, Kallam A, Vose JM, Kutateladze TG, El-Gamal D. A Novel Triple-Action Inhibitor Targeting B-Cell Receptor Signaling and BRD4 Demonstrates Preclinical Activity in Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:6712. [PMID: 35743155 PMCID: PMC9224275 DOI: 10.3390/ijms23126712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (CLL) results from intrinsic genetic defects and complex microenvironment stimuli that fuel CLL cell growth through an array of survival signaling pathways. Novel small-molecule agents targeting the B-cell receptor pathway and anti-apoptotic proteins alone or in combination have revolutionized the management of CLL, yet combination therapy carries significant toxicity and CLL remains incurable due to residual disease and relapse. Single-molecule inhibitors that can target multiple disease-driving factors are thus an attractive approach to combat both drug resistance and combination-therapy-related toxicities. We demonstrate that SRX3305, a novel small-molecule BTK/PI3K/BRD4 inhibitor that targets three distinctive facets of CLL biology, attenuates CLL cell proliferation and promotes apoptosis in a dose-dependent fashion. SRX3305 also inhibits the activation-induced proliferation of primary CLL cells in vitro and effectively blocks microenvironment-mediated survival signals, including stromal cell contact. Furthermore, SRX3305 blocks CLL cell migration toward CXCL-12 and CXCL-13, which are major chemokines involved in CLL cell homing and retention in microenvironment niches. Importantly, SRX3305 maintains its anti-tumor effects in ibrutinib-resistant CLL cells. Collectively, this study establishes the preclinical efficacy of SRX3305 in CLL, providing significant rationale for its development as a therapeutic agent for CLL and related disorders.
Collapse
Affiliation(s)
- Audrey L. Smith
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Alexandria P. Eiken
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Sydney A. Skupa
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Dalia Y. Moore
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Lelisse T. Umeta
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| | - Lynette M. Smith
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.M.S.); (E.R.L.)
| | - Elizabeth R. Lyden
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.M.S.); (E.R.L.)
| | - Christopher R. D’Angelo
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Avyakta Kallam
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Julie M. Vose
- Division of Hematology and Oncology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.R.D.); (A.K.); (J.M.V.)
| | - Tatiana G. Kutateladze
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Dalia El-Gamal
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (A.L.S.); (A.P.E.); (S.A.S.); (D.Y.M.); (L.T.U.)
| |
Collapse
|
7
|
Tang F, Xu D, Wang S, Wong CK, Martinez-Fundichely A, Lee CJ, Cohen S, Park J, Hill CE, Eng K, Bareja R, Han T, Liu EM, Palladino A, Di W, Gao D, Abida W, Beg S, Puca L, Meneses M, De Stanchina E, Berger MF, Gopalan A, Dow LE, Mosquera JM, Beltran H, Sternberg CN, Chi P, Scher HI, Sboner A, Chen Y, Khurana E. Chromatin profiles classify castration-resistant prostate cancers suggesting therapeutic targets. Science 2022; 376:eabe1505. [PMID: 35617398 PMCID: PMC9299269 DOI: 10.1126/science.abe1505] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In castration-resistant prostate cancer (CRPC), the loss of androgen receptor (AR) dependence leads to clinically aggressive tumors with few therapeutic options. We used ATAC-seq (assay for transposase-accessible chromatin sequencing), RNA-seq, and DNA sequencing to investigate 22 organoids, six patient-derived xenografts, and 12 cell lines. We identified the well-characterized AR-dependent and neuroendocrine subtypes, as well as two AR-negative/low groups: a Wnt-dependent subtype, and a stem cell-like (SCL) subtype driven by activator protein-1 (AP-1) transcription factors. We used transcriptomic signatures to classify 366 patients, which showed that SCL is the second most common subtype of CRPC after AR-dependent. Our data suggest that AP-1 interacts with the YAP/TAZ and TEAD proteins to maintain subtype-specific chromatin accessibility and transcriptomic landscapes in this group. Together, this molecular classification reveals drug targets and can potentially guide therapeutic decisions.
Collapse
Affiliation(s)
- Fanying Tang
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Shangqian Wang
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,State Key Laboratory of Reproductive Medicine, Urology department, the First Affiliated Hospital of Nanjing Medical University, Nanjing 211116, China
| | - Chen Khuan Wong
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alexander Martinez-Fundichely
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Cindy J. Lee
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sandra Cohen
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jane Park
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Corinne E. Hill
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kenneth Eng
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Rohan Bareja
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Teng Han
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric Minwei Liu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA.,Computational Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ann Palladino
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Wei Di
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dong Gao
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shaham Beg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Loredana Puca
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Maximiliano Meneses
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Elisa De Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michael F. Berger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anuradha Gopalan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lukas E. Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Juan Miguel Mosquera
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Himisha Beltran
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA.,Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Cora N. Sternberg
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA
| | - Howard I. Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Biomarker Development Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Sboner
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA.,Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Weill Cornell Medical College and New York-Presbyterian Hospital, New York, NY 10065, USA.,Corresponding authors. (E.K.); (Y.C.)
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY 10021, USA.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10021, USA.,Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021 USA.,Corresponding authors. (E.K.); (Y.C.)
| |
Collapse
|
8
|
Benoit A, Bou-Petit E, Chou H, Lu M, Guilbert C, Luo VM, Assouline S, Morin RD, Dmitrienko S, Estrada-Tejedor R, Johnson NA, Mann KK. Mutated RAS-associating proteins and ERK activation in relapse/refractory diffuse large B cell lymphoma. Sci Rep 2022; 12:779. [PMID: 35039569 PMCID: PMC8764096 DOI: 10.1038/s41598-021-04736-0] [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: 07/14/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is successfully treated with combination immuno-chemotherapy, but relapse with resistant disease occurs in ~ 40% of patients. However, little is known regarding relapsed/refractory DLBCL (rrDLBCL) genetics and alternative therapies. Based on findings from other tumors, we hypothesized that RAS-MEK-ERK signaling would be upregulated in resistant tumors, potentially correlating with mutations in RAS, RAF, or associated proteins. We analyzed mutations and phospho-ERK levels in tumor samples from rrDLBCL patients. Unlike other tumor types, rrDLBCL is not mutated in any Ras or Raf family members, despite having increased expression of p-ERK. In paired biopsies comparing diagnostic and relapsed specimens, 33% of tumors gained p-ERK expression, suggesting a role in promoting survival. We did find mutations in several Ras-associating proteins, including GEFs, GAPs, and downstream effectors that could account for increased ERK activation. We further investigated mutations in one such protein, RASGRP4. In silico modeling indicated an increased interaction between H-Ras and mutant RASGRP4. In cell lines, mutant RASGRP4 increased basal p-ERK expression and lead to a growth advantage in colony forming assays when challenged with doxorubicin. Relapsed/refractory DLBCL is often associated with increased survival signals downstream of ERK, potentially corresponding with mutations in protein controlling RAS/MEK/ERK signaling.
Collapse
Affiliation(s)
- Alexandre Benoit
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Elisabeth Bou-Petit
- Grup de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - Hsiang Chou
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Melissa Lu
- Université de Montréal-Faculté de Médecine, Montreal, Canada
| | - Cynthia Guilbert
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada
| | - Vincent Mingyi Luo
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Sarit Assouline
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Ryan D Morin
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Svetlana Dmitrienko
- Division of Pathology, McGill University Health Centre, Montreal, QC, Canada
| | - Roger Estrada-Tejedor
- Grup de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Barcelona, Spain
| | - Nathalie A Johnson
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Koren K Mann
- Lady Davis Institute, Jewish General Hospital, 3755 Côte Sainte-Catherine Road, Montreal, QC, H3T 1E2, Canada. .,Division of Experimental Medicine, McGill University, Montreal, QC, Canada. .,Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
Penter L, Gohil SH, Wu CJ. Natural Barcodes for Longitudinal Single Cell Tracking of Leukemic and Immune Cell Dynamics. Front Immunol 2022; 12:788891. [PMID: 35046946 PMCID: PMC8761982 DOI: 10.3389/fimmu.2021.788891] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/08/2021] [Indexed: 11/26/2022] Open
Abstract
Blood malignancies provide unique opportunities for longitudinal tracking of disease evolution following therapeutic bottlenecks and for the monitoring of changes in anti-tumor immunity. The expanding development of multi-modal single-cell sequencing technologies affords newer platforms to elucidate the mechanisms underlying these processes at unprecedented resolution. Furthermore, the identification of molecular events that can serve as in-vivo barcodes now facilitate the tracking of the trajectories of malignant and of immune cell populations over time within primary human samples, as these permit unambiguous identification of the clonal lineage of cell populations within heterogeneous phenotypes. Here, we provide an overview of the potential for chromosomal copy number changes, somatic nuclear and mitochondrial DNA mutations, single nucleotide polymorphisms, and T and B cell receptor sequences to serve as personal natural barcodes and review technical implementations in single-cell analysis workflows. Applications of these methodologies include the study of acquired therapeutic resistance and the dissection of donor- and host cellular interactions in the context of allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Satyen H. Gohil
- Department of Academic Haematology, University College London Cancer Institute, London, United Kingdom
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
- Harvard Medical School, Boston, MA, United States
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
10
|
Kambhampati S, Song JY, Herrera AF, Chan WC. Barriers to achieving a cure in lymphoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:965-983. [PMID: 35582375 PMCID: PMC8992454 DOI: 10.20517/cdr.2021.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/08/2021] [Accepted: 10/26/2021] [Indexed: 11/23/2022]
Abstract
Lymphoma is a diverse disease with a variety of different subtypes, each characterized by unique pathophysiology, tumor microenvironment, and underlying signaling pathways leading to oncogenesis. With our increasing understanding of the molecular biology of lymphoma, there have been a number of novel targeted therapies and immunotherapy approaches that have been developed for the treatment of this complex disease. Despite rapid progress in the field, however, many patients still relapse largely due to the development of drug resistance to these therapies. A better understanding of the mechanisms underlying resistance is needed to develop more novel treatment strategies that circumvent these mechanisms and design better treatment algorithms that personalize therapies to patients and sequence these therapies in the most optimal manner. This review focuses on the recent advances in therapies in lymphoma, including targeted therapies, monoclonal antibodies, antibody-drug conjugates, cellular therapy, bispecific antibodies, and checkpoint inhibitors. We discuss the genetic and cellular principles of drug resistance that span across all the therapies, as well as some of the unique mechanisms of resistance that are specific to these individual classes of therapies and the strategies that have been developed to address these modes of resistance.
Collapse
Affiliation(s)
- Swetha Kambhampati
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Joo Y. Song
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Alex F. Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
11
|
Genetic Events Inhibiting Apoptosis in Diffuse Large B Cell Lymphoma. Cancers (Basel) 2021; 13:cancers13092167. [PMID: 33946435 PMCID: PMC8125500 DOI: 10.3390/cancers13092167] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma (NHL). Despite the genetic heterogeneity of the disease, most patients are initially treated with a combination of rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP), but relapse occurs in ~50% of patients. One of the hallmarks of DLBCL is the occurrence of genetic events that inhibit apoptosis, which contributes to disease development and resistance to therapy. These events can affect the intrinsic or extrinsic apoptotic pathways, or their modulators. Understanding the factors that contribute to inhibition of apoptosis in DLBCL is crucial in order to be able to develop targeted therapies and improve outcomes, particularly in relapsed and refractory DLBCL (rrDLBCL). This review provides a description of the genetic events inhibiting apoptosis in DLBCL, their contribution to lymphomagenesis and chemoresistance, and their implication for the future of DLBCL therapy. Abstract Diffuse large B cell lymphoma (DLBCL) is curable with chemoimmunotherapy in ~65% of patients. One of the hallmarks of the pathogenesis and resistance to therapy in DLBCL is inhibition of apoptosis, which allows malignant cells to survive and acquire further alterations. Inhibition of apoptosis can be the result of genetic events inhibiting the intrinsic or extrinsic apoptotic pathways, as well as their modulators, such as the inhibitor of apoptosis proteins, P53, and components of the NF-kB pathway. Mechanisms of dysregulation include upregulation of anti-apoptotic proteins and downregulation of pro-apoptotic proteins via point mutations, amplifications, deletions, translocations, and influences of other proteins. Understanding the factors contributing to resistance to apoptosis in DLBCL is crucial in order to be able to develop targeted therapies that could improve outcomes by restoring apoptosis in malignant cells. This review describes the genetic events inhibiting apoptosis in DLBCL, provides a perspective of their interactions in lymphomagenesis, and discusses their implication for the future of DLBCL therapy.
Collapse
|
12
|
Lee B, Lee H, Cho J, Yoon SE, Kim SJ, Park WY, Kim WS, Ko YH. Mutational Profile and Clonal Evolution of Relapsed/Refractory Diffuse Large B-Cell Lymphoma. Front Oncol 2021; 11:628807. [PMID: 33777778 PMCID: PMC7992425 DOI: 10.3389/fonc.2021.628807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Primary refractory/relapsed diffuse large B-cell lymphoma (rrDLBCL) is an unresolved issue for DLBCL treatment and new treatments to overcome resistance is required. To explore the genetic mechanisms underlying treatment resistance in rrDLBCL and to identify candidate genes, we performed targeted deep sequencing of 430 lymphoma-related genes from 58 patients diagnosed with rrDLBCL. Genetic alterations found between the initial biopsy and biopsy at recurrence or refractory disease were investigated. The genes most frequently altered (> 20%) were (in decreasing order of frequency) CDKN2A, PIM1, CD79B, TP53, MYD88, MYC, BTG2, BTG1, CDKN2B, DTX1, CD58, ETV6, and IRF4. Genes mutation of which in pretreatment sample were associated with poor overall survival included NOTCH1, FGFR2, BCL7A, BCL10, SPEN and TP53 (P < 0.05). FGFR2, BCL2, BCL6, BCL10, and TP53 were associated with poor progression-free survival (P < 0.05). Most mutations were truncal and were maintained in both the initial biopsy and post-treatment biopsy with high dynamics of subclones. Immune-evasion genes showed increased overall mutation frequency (CD58, B2M) and variant allele fraction (CD58), and decreased copy number (B2M, CD70) at the post-treatment biopsy. Using the established mutational profiles and integrative analysis of mutational evolution, we identified information about candidate genes that may be useful for the development of future treatment strategies.
Collapse
Affiliation(s)
- Boram Lee
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hyunwoo Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Junhun Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang Eun Yoon
- Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seok Jin Kim
- Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Won Seog Kim
- Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Hyeh Ko
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
13
|
Bakhshi TJ, Georgel PT. Genetic and epigenetic determinants of diffuse large B-cell lymphoma. Blood Cancer J 2020; 10:123. [PMID: 33277464 PMCID: PMC7718920 DOI: 10.1038/s41408-020-00389-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/25/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common type of lymphoma and is notorious for its heterogeneity, aggressive nature, and the frequent development of resistance and/or relapse after treatment with standard chemotherapy. To address these problems, a strong emphasis has been placed on researching the molecular origins and mechanisms of DLBCL to develop effective treatments. One of the major insights produced by such research is that DLBCL almost always stems from genetic damage that occurs during the germinal center (GC) reaction, which is required for the production of high-affinity antibodies. Indeed, there is significant overlap between the mechanisms that govern the GC reaction and those that drive the progression of DLBCL. A second important insight is that some of the most frequent genetic mutations that occur in DLBCL are those related to chromatin and epigenetics, especially those related to proteins that “write” histone post-translational modifications (PTMs). Mutation or deletion of these epigenetic writers often renders cells unable to epigenetically “switch on” critical gene sets that are required to exit the GC reaction, differentiate, repair DNA, and other essential cellular functions. Failure to activate these genes locks cells into a genotoxic state that is conducive to oncogenesis and/or relapse.
Collapse
Affiliation(s)
- Tanner J Bakhshi
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Philippe T Georgel
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA. .,Department of Biological Sciences, Cell Differentiation and Development Center, Byrd Biotechnology Science Center, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
14
|
Berendsen MR, Stevens WBC, van den Brand M, van Krieken JH, Scheijen B. Molecular Genetics of Relapsed Diffuse Large B-Cell Lymphoma: Insight into Mechanisms of Therapy Resistance. Cancers (Basel) 2020; 12:E3553. [PMID: 33260693 PMCID: PMC7760867 DOI: 10.3390/cancers12123553] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022] Open
Abstract
The majority of patients with diffuse large B-cell lymphoma (DLBCL) can be treated successfully with a combination of chemotherapy and the monoclonal anti-CD20 antibody rituximab. Nonetheless, approximately one-third of the patients with DLBCL still experience relapse or refractory (R/R) disease after first-line immunochemotherapy. Whole-exome sequencing on large cohorts of primary DLBCL has revealed the mutational landscape of DLBCL, which has provided a framework to define novel prognostic subtypes in DLBCL. Several studies have investigated the genetic alterations specifically associated with R/R DLBCL, thereby uncovering molecular pathways linked to therapy resistance. Here, we summarize the current state of knowledge regarding the genetic alterations that are enriched in R/R DLBCL, and the corresponding pathways affected by these gene mutations. Furthermore, we elaborate on their potential role in mediating therapy resistance, also in connection with findings in other B-cell malignancies, and discuss alternative treatment options. Hence, this review provides a comprehensive overview on the gene lesions and molecular mechanisms underlying R/R DLBCL, which are considered valuable parameters to guide treatment.
Collapse
Affiliation(s)
- Madeleine R. Berendsen
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
- Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| | - Wendy B. C. Stevens
- Department of Hematology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands;
| | - Michiel van den Brand
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
- Pathology-DNA, Rijnstate Hospital, 6815AD Arnhem, The Netherlands
| | - J. Han van Krieken
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
| | - Blanca Scheijen
- Department of Pathology, Radboud University Medical Center, 6525GA Nijmegen, The Netherlands; (M.R.B.); (M.v.d.B.); (J.H.v.K.)
- Radboud Institute for Molecular Life Sciences, 6525GA Nijmegen, The Netherlands
| |
Collapse
|
15
|
Genetic heterogeneity and prognostic impact of recurrent ANK2 and TP53 mutations in mantle cell lymphoma: a multi-centre cohort study. Sci Rep 2020; 10:13359. [PMID: 32770099 PMCID: PMC7414214 DOI: 10.1038/s41598-020-70310-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
The molecular features of mantle cell lymphoma (MCL), including its increased incidence, and complex therapies have not been investigated in detail, particularly in East Asian populations. In this study, we performed targeted panel sequencing (TPS) and whole-exome sequencing (WES) to investigate the genetic alterations in Korean MCL patients. We obtained a total of 53 samples from MCL patients from five Korean university hospitals between 2009 and 2016. We identified the recurrently mutated genes such as SYNE1, ATM, KMT2D, CARD11, ANK2, KMT2C, and TP53, which included some known drivers of MCL. The mutational profiles of our cohort indicated genetic heterogeneity. The significantly enriched pathways were mainly involved in gene expression, cell cycle, and programmed cell death. Multivariate analysis revealed that ANK2 mutations impacted the unfavourable overall survival (hazard ratio [HR] 3.126; P = 0.032). Furthermore, TP53 mutations were related to worse progression-free survival (HR 7.813; P = 0.043). Among the recurrently mutated genes with more than 15.0% frequency, discrepancies were found in only 5 genes from 4 patients, suggesting comparability of the TPS to WES in practical laboratory settings. We provide the unbiased genetic landscape that might contribute to MCL pathogenesis and recurrent genes conferring unfavourable outcomes.
Collapse
|
16
|
Wise JF, Nakken S, Steen CB, Vodák D, Trøen G, Johannessen B, Lingjærde OC, Hilden V, Blaker YN, Bai B, Aasheim LB, Pasanen A, Lorenz S, Sveen A, Lothe RA, Myklebost O, Leppä S, Meza-Zepeda LA, Beiske K, Lawrence MS, Hovig E, Myklebust JH, Smeland EB, Holte H. Mutational dynamics and immune evasion in diffuse large B-cell lymphoma explored in a relapse-enriched patient series. Blood Adv 2020; 4:1859-1866. [PMID: 32374878 PMCID: PMC7218413 DOI: 10.1182/bloodadvances.2019001325] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/18/2020] [Indexed: 12/31/2022] Open
Abstract
Diagnostic and relapse diffuse large B-cell lymphoma (DLBCL) biopsies reveal increased mutational burden/loss of heterozygosity in HLA-A . Serially sampled tumor biopsies provide insight into therapeutic targets and evolutionary divergence in relapsed/refractory DLBCL.
Collapse
Affiliation(s)
- Jillian F Wise
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
- Massachusetts General Hospital Cancer Center, Boston, MA
- Department of Pathology, Harvard Medical School, Charlestown, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Sigve Nakken
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, and
| | - Chloé B Steen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Daniel Vodák
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine
| | | | - Bjarne Johannessen
- Institute of Clinical Medicine, Faculty of Medicine
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | - Vera Hilden
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Yngvild Nuvin Blaker
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine
| | - Baoyan Bai
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Lars Birger Aasheim
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Annika Pasanen
- Department of Oncology, Helsinki University Hospital Cancer Center, Helsinki, Finland
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Susanne Lorenz
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Genomics Core Facility, Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Anita Sveen
- Institute of Clinical Medicine, Faculty of Medicine
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A Lothe
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ola Myklebost
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department for Clinical Science, University of Bergen, Bergen, Norway; and
| | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital Cancer Center, Helsinki, Finland
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Leonardo A Meza-Zepeda
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Genomics Core Facility, Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Klaus Beiske
- Institute of Clinical Medicine, Faculty of Medicine
- Department of Pathology and
| | - Michael S Lawrence
- Massachusetts General Hospital Cancer Center, Boston, MA
- Department of Pathology, Harvard Medical School, Charlestown, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
| | - Eivind Hovig
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - June Helen Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Erlend B Smeland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Harald Holte
- Norwegian Cancer Genomics Consortium, Oslo, Norway
- K. G. Jebsen Centre for B-cell Malignancies, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
17
|
Drug Resistance in Non-Hodgkin Lymphomas. Int J Mol Sci 2020; 21:ijms21062081. [PMID: 32197371 PMCID: PMC7139754 DOI: 10.3390/ijms21062081] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
Non-Hodgkin lymphomas (NHL) are lymphoid tumors that arise by a complex process of malignant transformation of mature lymphocytes during various stages of differentiation. The WHO classification of NHL recognizes more than 90 nosological units with peculiar pathophysiology and prognosis. Since the end of the 20th century, our increasing knowledge of the molecular biology of lymphoma subtypes led to the identification of novel druggable targets and subsequent testing and clinical approval of novel anti-lymphoma agents, which translated into significant improvement of patients’ outcome. Despite immense progress, our effort to control or even eradicate malignant lymphoma clones has been frequently hampered by the development of drug resistance with ensuing unmet medical need to cope with relapsed or treatment-refractory disease. A better understanding of the molecular mechanisms that underlie inherent or acquired drug resistance might lead to the design of more effective front-line treatment algorithms based on reliable predictive markers or personalized salvage therapy, tailored to overcome resistant clones, by targeting weak spots of lymphoma cells resistant to previous line(s) of therapy. This review focuses on the history and recent advances in our understanding of molecular mechanisms of resistance to genotoxic and targeted agents used in clinical practice for the therapy of NHL.
Collapse
|
18
|
Odabashian M, Carlotti E, Araf S, Okosun J, Spada F, Gribben JG, Forconi F, Stevenson FK, Calaminici M, Krysov S. IGHV sequencing reveals acquired N-glycosylation sites as a clonal and stable event during follicular lymphoma evolution. Blood 2020; 135:834-844. [PMID: 31932843 PMCID: PMC7195541 DOI: 10.1182/blood.2019002279] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/13/2019] [Indexed: 12/24/2022] Open
Abstract
Follicular lymphoma B cells undergo continuous somatic hypermutation (SHM) of their immunoglobulin variable region genes, generating a heterogeneous tumor population. SHM introduces DNA sequences encoding N-glycosylation sites asparagine-X-serine/threonine (N-gly sites) within the V-region that are rarely found in normal B-cell counterparts. Unique attached oligomannoses activate B-cell receptor signaling pathways after engagement with calcium-dependent lectins expressed by tissue macrophages. This novel interaction appears critical for tumor growth and survival. To elucidate the significance of N-gly site presence and loss during ongoing SHM, we tracked site behavior during tumor evolution and progression in a diverse group of patients through next-generation sequencing. A hierarchy of subclones was visualized through lineage trees based on SHM semblance between subclones and their discordance from the germline sequence. We observed conservation of N-gly sites in more than 96% of subclone populations within and across diagnostic, progression, and transformation events. Rare N-gly-negative subclones were lost or negligible from successive events, in contrast to N-gly-positive subclones, which could additionally migrate between anatomical sites. Ongoing SHM of the N-gly sites resulted in subclones with different amino acid compositions across disease events, yet the vast majority of resulting DNA sequences still encoded for an N-gly site. The selection and expansion of only N-gly-positive subclones is evidence of the tumor cells' dependence on sites, despite the changing genomic complexity as the disease progresses. N-gly sites were gained in the earliest identified lymphoma cells, indicating they are an early and stable event of pathogenesis. Targeting the inferred mannose-lectin interaction holds therapeutic promise.
Collapse
Affiliation(s)
- Mariette Odabashian
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Emanuela Carlotti
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Shamzah Araf
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Filomena Spada
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - John G Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Francesco Forconi
- Cancer Sciences Division, Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
| | - Freda K Stevenson
- Cancer Sciences Division, Somers Cancer Sciences Building, University of Southampton, Southampton, United Kingdom
| | - Mariarita Calaminici
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| | - Sergey Krysov
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom; and
| |
Collapse
|
19
|
Molecular Complexity of Diffuse Large B-Cell Lymphoma: Can It Be a Roadmap for Precision Medicine? Cancers (Basel) 2020; 12:cancers12010185. [PMID: 31940809 PMCID: PMC7017344 DOI: 10.3390/cancers12010185] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/11/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma; it features extreme molecular heterogeneity regardless of the classical cell-of-origin (COO) classification. Despite this, the standard therapeutic approach is still immunochemotherapy (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone-R-CHOP), which allows a 60% overall survival (OS) rate, but up to 40% of patients experience relapse or refractory (R/R) disease. With the purpose of searching for new clinical parameters and biomarkers helping to make a better DLBCL patient characterization and stratification, in the last years a series of large discovery genomic and transcriptomic studies has been conducted, generating a wealth of information that needs to be put in order. We reviewed these researches, trying ultimately to understand if there are bases offering a roadmap toward personalized and precision medicine also for DLBCL.
Collapse
|
20
|
Bewicke-Copley F, Arjun Kumar E, Palladino G, Korfi K, Wang J. Applications and analysis of targeted genomic sequencing in cancer studies. Comput Struct Biotechnol J 2019; 17:1348-1359. [PMID: 31762958 PMCID: PMC6861594 DOI: 10.1016/j.csbj.2019.10.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 12/31/2022] Open
Abstract
Next Generation Sequencing (NGS) has dramatically improved the flexibility and outcomes of cancer research and clinical trials, providing highly sensitive and accurate high-throughput platforms for large-scale genomic testing. In contrast to whole-genome (WGS) or whole-exome sequencing (WES), targeted genomic sequencing (TS) focuses on a panel of genes or targets known to have strong associations with pathogenesis of disease and/or clinical relevance, offering greater sequencing depth with reduced costs and data burden. This allows targeted sequencing to identify low frequency variants in targeted regions with high confidence, thus suitable for profiling low-quality and fragmented clinical DNA samples. As a result, TS has been widely used in clinical research and trials for patient stratification and the development of targeted therapeutics. However, its transition to routine clinical use has been slow. Many technical and analytical obstacles still remain and need to be discussed and addressed before large-scale and cross-centre implementation. Gold-standard and state-of-the-art procedures and pipelines are urgently needed to accelerate this transition. In this review we first present how TS is conducted in cancer research, including various target enrichment platforms, the construction of target panels, and selected research and clinical studies utilising TS to profile clinical samples. We then present a generalised analytical workflow for TS data discussing important parameters and filters in detail, aiming to provide the best practices of TS usage and analyses.
Collapse
Key Words
- BAM, Binary Alignment Map
- BWA, Burrows-Wheeler Aligner
- Background error
- CLL, Chronic Lymphocytic Leukaemia
- COSMIC, Catalogue of Somatic Mutations in Cancer
- Cancer genomics
- Clinical samples
- ESP, Exome Sequencing Project
- FF, Fresh Frozen
- FFPE, Formalin Fixed Paraffin Embedded
- FL, Follicular Lymphoma
- GATK, Genome Analysis Toolkit
- ICGC, International Cancer Genome Consortium
- MBC, Molecular Barcode
- NCCN, the National Comprehensive Cancer Network®
- NGS, Next Generation Sequencing
- NHL, Non-Hodgkin Lymphoma
- NSCLC, Non-Small Cell Lung Carcinoma
- PCR duplicates
- QC, Quality Control
- SAM, Sequence Alignment Map
- TCGA, The Cancer Genome Atlas
- TS, Targeted Sequencing
- Targeted sequencing
- UMI, Unique Molecular Identifiers
- VAF, Variant Allele Frequency
- Variant calling
- WES, Whole Exome Sequencing
- WGS, Whole Genome Sequencing
- tFL, Transformed Follicular Lymphoma
Collapse
Affiliation(s)
- Findlay Bewicke-Copley
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Emil Arjun Kumar
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Giuseppe Palladino
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Koorosh Korfi
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jun Wang
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
21
|
Ma J, Redmond D, Miyaguchi A, Nam AS, Nie K, Mathew S, Elemento O, Tam W. Exploring tumor clonal evolution in bone marrow of patients with diffuse large B-cell lymphoma by deep IGH sequencing and its potential relevance in relapse. Blood Cancer J 2019; 9:69. [PMID: 31434873 PMCID: PMC6704167 DOI: 10.1038/s41408-019-0229-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/13/2019] [Accepted: 05/24/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Jiao Ma
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiaotong University, School of Medicine, Shanghai, China.,Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David Redmond
- Department of Physiology and Biophysics & Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Ayako Miyaguchi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Anna S Nam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kui Nie
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Susan Mathew
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics & Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
22
|
Barranco GI, Fernández S, Oña R, González-Rincón J, Martínez-Ramírez A, Teijo A, Camacho FI, Pinedo FJ, Sánchez-Beato M, Pedrosa L, de la Fuente A, Estévez M, Iglesias R, Montalbán C, García JF. Branched clonal evolution: nodal follicular lymphoma and primary diffuse large B-cell lymphoma of the central nervous system. Haematologica 2019; 104:e326-e329. [PMID: 30890596 DOI: 10.3324/haematol.2018.214981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Gilberto I Barranco
- Hospital General de México, Instituto Nacional de Cancerología, Ciudad de México, Mexico.,Department of Pathology, MD Anderson Cancer Center Madrid, Centro de Investigación Biomédica en Red de Oncología (CIBERONC-ISCIII), Madrid, Spain
| | - Sara Fernández
- Translational Research, MD Anderson Cancer Center Madrid, CIBERONC-ISCI-II, Madrid, Spain
| | - Raquel Oña
- Hematology, MD Anderson Cancer Center Madrid, Spain
| | - Julia González-Rincón
- Lymphoma Research Group, Medical Oncology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), CIBERONC-ISCIII, Madrid, Spain
| | | | - Ana Teijo
- Hospital General de México, Instituto Nacional de Cancerología, Ciudad de México, Mexico
| | | | | | - Margarita Sánchez-Beato
- Lymphoma Research Group, Medical Oncology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), CIBERONC-ISCIII, Madrid, Spain
| | - Lucia Pedrosa
- Lymphoma Research Group, Medical Oncology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana (IDIPHISA), CIBERONC-ISCIII, Madrid, Spain
| | | | | | | | | | - Juan F García
- Hospital General de México, Instituto Nacional de Cancerología, Ciudad de México, Mexico .,Translational Research, MD Anderson Cancer Center Madrid, CIBERONC-ISCI-II, Madrid, Spain
| |
Collapse
|
23
|
Mutational Evolution in Relapsed Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2018; 10:cancers10110459. [PMID: 30463380 PMCID: PMC6265691 DOI: 10.3390/cancers10110459] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
Current genomic models in diffuse large B-cell lymphoma (DLBCL) are based on single tumor biopsies, which might underestimate heterogeneity. Data on mutational evolution largely remains unknown. An exploratory study using whole exome sequencing on paired (primary and relapse) formalin fixed paraffin embedded DLBCL biopsies (n = 14) of 6 patients was performed to globally assess the mutational evolution and to identify gene mutations specific for relapse samples from patients treated with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisolone. A minority of the mutations detected in the primary sample (median 7.6%, range 4.8–66.2%) could not be detected in the matching relapse sample. Relapsed DLBCL samples showed a mild increase of mutations (median 12.5%, range 9.4–87.6%) as compared to primary tumor biopsies. We identified 264 genes possibly related to therapy resistance, including tyrosine kinases (n = 18), (transmembrane) glycoproteins (n = 73), and genes involved in the JAK-STAT pathway (n = 7). Among the potentially resistance related genes were PIM1, SOCS1, and MYC, which have been reported to convey a risk for treatment failure. In conclusion, we show modest temporal heterogeneity between paired tumor samples with the acquisition of new mutations and identification of genes possibly related to therapy resistance. The mutational evolution could have implications for treatment decisions and development of novel targeted drugs.
Collapse
|
24
|
Circulating tumor DNA analysis depicts subclonal architecture and genomic evolution of small cell lung cancer. Nat Commun 2018; 9:3114. [PMID: 30082701 PMCID: PMC6079068 DOI: 10.1038/s41467-018-05327-w] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/29/2018] [Indexed: 11/10/2022] Open
Abstract
Subclonal architecture and genomic evolution of small-cell lung cancer (SCLC) under treatment has not been well studied primarily due to lack of tumor specimens, particularly longitudinal samples acquired during treatment. SCLC is characterized by early hematogenous spread, which makes circulating cell-free tumor DNA (ctDNA) sequencing a promising modality for genomic profiling. Here, we perform targeted deep sequencing of 430 cancer genes on pre-treatment tumor biopsies, as well as on plasma samples collected prior to and during treatment from 22 SCLC patients. Similar subclonal architecture is observed between pre-treatment ctDNA and paired tumor DNA. Mean variant allele frequency of clonal mutations from pre-treatment ctDNA is associated with progression-free survival and overall survival. Pre- and post-treatment ctDNA mutational analysis demonstrate that mutations of DNA repair and NOTCH signaling pathways are enriched in post-treatment samples. These data suggest that ctDNA sequencing is promising to delineate genomic landscape, subclonal architecture, and genomic evolution of SCLC. Small cell lung cancer (SCLC) may evolve under treatment. But tumor tissues are often not available to study evolution of SCLC. Here, the authors utilize circulating tumor DNA to investigate the genomic evolution and subclonal architecture of SCLC during therapy.
Collapse
|
25
|
Extracellular vesicles in DLBCL provide abundant clues to aberrant transcriptional programming and genomic alterations. Blood 2018; 132:e13-e23. [PMID: 29967128 DOI: 10.1182/blood-2017-12-821843] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/21/2018] [Indexed: 12/16/2022] Open
Abstract
The biological role of extracellular vesicles (EVs) in diffuse large B-cell lymphoma (DLBCL) initiation and progression remains largely unknown. We characterized EVs secreted by 5 DLBCL cell lines, a primary DLBCL tumor, and a normal control B-cell sample, optimized their purification, and analyzed their content. We found that DLBCLs secreted large quantities of CD63, Alix, TSG101, and CD81 EVs, which can be extracted using an ultracentrifugation-based method and traced by their cell of origin surface markers. We also showed that tumor-derived EVs can be exchanged between lymphoma cells, normal tonsillar cells, and HK stromal cells. We then examined the content of EVs, focusing on isolation of high-quality total RNA. We sequenced the total RNA and analyzed the nature of RNA species, including coding and noncoding RNAs. We compared whole-cell and EV-derived RNA composition in benign and malignant B cells and discovered that transcripts from EVs were involved in many critical cellular functions. Finally, we performed mutational analysis and found that mutations detected in EVs exquisitely represented mutations in the cell of origin. These results enhance our understanding and enable future studies of the role that EVs may play in the pathogenesis of DLBCL, particularly with regards to the exchange of genomic information. Current findings open a new strategy for liquid biopsy approaches in disease monitoring.
Collapse
|
26
|
Pasqualucci L, Dalla-Favera R. Genetics of diffuse large B-cell lymphoma. Blood 2018; 131:2307-2319. [PMID: 29666115 PMCID: PMC5969374 DOI: 10.1182/blood-2017-11-764332] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/15/2018] [Indexed: 02/07/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL), the most frequent subtype of lymphoid malignancy, remains a significant clinical challenge, as ∼30% of patients are not cured. Over the past decade, remarkable progress has been made in the understanding of the pathogenesis of this disease, spurred by the implementation of powerful genomic technologies that enabled the definition of its genetic and epigenetic landscape. These studies have uncovered a multitude of genomic alterations that contribute to the initiation and maintenance of the tumor clone by disrupting biological functions known to be critical for the normal biology of its cells of origin, germinal center B cells. The identified alterations involve epigenetic remodeling, block of differentiation, escape from immune surveillance, and the constitutive activation of several signal transduction pathways. This wealth of new information offers unique opportunities for the development of improved diagnostic and prognostic tools that could help guide the clinical management of DLBCL patients. Furthermore, a number of the mutated genes identified are potentially actionable targets that are currently being explored for the development of novel therapeutic strategies. This review summarizes current knowledge of the most common genetic alterations associated with DLBCL in relation to their functional impact on the malignant transformation process, and discusses their clinical implications for mechanism-based therapeutics.
Collapse
Affiliation(s)
- Laura Pasqualucci
- Institute for Cancer Genetics
- Department of Pathology and Cell Biology
| | - Riccardo Dalla-Favera
- Institute for Cancer Genetics
- Department of Pathology and Cell Biology
- Department of Genetics, and
- Department of Microbiology and Immunology, Columbia University, New York, NY
| |
Collapse
|
27
|
Ozer HG, El-Gamal D, Powell B, Hing ZA, Blachly JS, Harrington B, Mitchell S, Grieselhuber NR, Williams K, Lai TH, Alinari L, Baiocchi RA, Brinton L, Baskin E, Cannon M, Beaver L, Goettl VM, Lucas DM, Woyach JA, Sampath D, Lehman AM, Yu L, Zhang J, Ma Y, Zhang Y, Spevak W, Shi S, Severson P, Shellooe R, Carias H, Tsang G, Dong K, Ewing T, Marimuthu A, Tantoy C, Walters J, Sanftner L, Rezaei H, Nespi M, Matusow B, Habets G, Ibrahim P, Zhang C, Mathé EA, Bollag G, Byrd JC, Lapalombella R. BRD4 Profiling Identifies Critical Chronic Lymphocytic Leukemia Oncogenic Circuits and Reveals Sensitivity to PLX51107, a Novel Structurally Distinct BET Inhibitor. Cancer Discov 2018; 8:458-477. [PMID: 29386193 PMCID: PMC5882533 DOI: 10.1158/2159-8290.cd-17-0902] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 12/12/2017] [Accepted: 01/26/2018] [Indexed: 11/16/2022]
Abstract
Bromodomain and extra-terminal (BET) family proteins are key regulators of gene expression in cancer. Herein, we utilize BRD4 profiling to identify critical pathways involved in pathogenesis of chronic lymphocytic leukemia (CLL). BRD4 is overexpressed in CLL and is enriched proximal to genes upregulated or de novo expressed in CLL with known functions in disease pathogenesis and progression. These genes, including key members of the B-cell receptor (BCR) signaling pathway, provide a rationale for this therapeutic approach to identify new targets in alternative types of cancer. Additionally, we describe PLX51107, a structurally distinct BET inhibitor with novel in vitro and in vivo pharmacologic properties that emulates or exceeds the efficacy of BCR signaling agents in preclinical models of CLL. Herein, the discovery of the involvement of BRD4 in the core CLL transcriptional program provides a compelling rationale for clinical investigation of PLX51107 as epigenetic therapy in CLL and application of BRD4 profiling in other cancers.Significance: To date, functional studies of BRD4 in CLL are lacking. Through integrated genomic, functional, and pharmacologic analyses, we uncover the existence of BRD4-regulated core CLL transcriptional programs and present preclinical proof-of-concept studies validating BET inhibition as an epigenetic approach to target BCR signaling in CLL. Cancer Discov; 8(4); 458-77. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 371.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Cycle Proteins
- Cell Line, Tumor
- Cell Proliferation
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Humans
- Isoxazoles/pharmacology
- Isoxazoles/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology
- Mice
- Mice, SCID
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Pyridines/pharmacology
- Pyridines/therapeutic use
- Pyrroles/pharmacology
- Pyrroles/therapeutic use
- Signal Transduction
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Hatice Gulcin Ozer
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Dalia El-Gamal
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | | | - Zachary A Hing
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - James S Blachly
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Bonnie Harrington
- College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Shaneice Mitchell
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Nicole R Grieselhuber
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Katie Williams
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Tzung-Huei Lai
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Lapo Alinari
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Robert A Baiocchi
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Lindsey Brinton
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Elizabeth Baskin
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Matthew Cannon
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Larry Beaver
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Virginia M Goettl
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - David M Lucas
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Jennifer A Woyach
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Deepa Sampath
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio
| | - Amy M Lehman
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | - Lianbo Yu
- Center for Biostatistics, The Ohio State University, Columbus, Ohio
| | | | - Yan Ma
- Plexxikon Inc., Berkeley, California
| | | | | | | | | | | | | | | | - Ken Dong
- Plexxikon Inc., Berkeley, California
| | | | | | | | | | | | | | | | | | | | | | | | - Ewy A Mathé
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | | | - John C Byrd
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio.
| | - Rosa Lapalombella
- Division of Hematology, Department of Medicine, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
28
|
Clonal evolution in relapsed and refractory diffuse large B-cell lymphoma is characterized by high dynamics of subclones. Oncotarget 2018; 7:51494-51502. [PMID: 27285986 PMCID: PMC5239491 DOI: 10.18632/oncotarget.9860] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 05/22/2016] [Indexed: 11/25/2022] Open
Abstract
Little information is available about the role of certain mutations for clonal evolution and the clinical outcome during relapse in diffuse large B-cell lymphoma (DLBCL). Therefore, we analyzed formalin-fixed-paraffin-embedded tumor samples from first diagnosis, relapsed or refractory disease from 28 patients using next-generation sequencing of the exons of 104 coding genes. Non-synonymous mutations were present in 74 of the 104 genes tested. Primary tumor samples showed a median of 8 non-synonymous mutations (range: 0-24) with the used gene set. Lower numbers of non-synonymous mutations in the primary tumor were associated with a better median OS compared with higher numbers (28 versus 15 months, p=0.031). We observed three patterns of clonal evolution during relapse of disease: large global change, subclonal selection and no or minimal change possibly suggesting preprogrammed resistance. We conclude that targeted re-sequencing is a feasible and informative approach to characterize the molecular pattern of relapse and it creates novel insights into the role of dynamics of individual genes.
Collapse
|
29
|
Lee SE, Kang SY, Yoo HY, Kim SJ, Kim WS, Ko YH. Clonal relationships in recurrent B-cell lymphomas. Oncotarget 2017; 7:12359-71. [PMID: 26848863 PMCID: PMC4914290 DOI: 10.18632/oncotarget.7132] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/19/2016] [Indexed: 11/25/2022] Open
Abstract
Immunoglobulin (Ig) gene rearrangements remain largely unmodified during the clonal expansion of neoplastic cells. We investigated the clonal relationships between lymphoma components at diagnosis and at relapse by analyzing Ig gene rearrangements. A BIOMED-2 multiplex polymerase chain reaction (PCR) assay was performed in 27 patients using formalin-fixed paraffin embedded tissues, with subsequent cloning and sequencing of the amplified Ig genes in 17 patients. All 27 cases of primary and corresponding relapsed tumors showed monoclonal rearrangements of the Ig genes by BIOMED-2 PCR. Whereas IgVH or IgVK fragment lengths were identical in 8/27 pairs (30%), fragment lengths differed in 19/27 pairs (70%). In 17 cases analyzed by sequencing, an identical VDJ gene rearrangement was confirmed in 4/4 pairs (100%) with the same fragment lengths and in 10/13 pairs (77%) with different fragment lengths. Four of 17 primary lymphomas had multiple VDJ rearrangements, and three of them showed an unrelated relapse. Unrelated relapse was observed in 1/8 mantle cell lymphomas, 1/5 diffuse large B-cell lymphomas, and a large B cell lymphoma developed in a patient with a small lymphocytic lymphoma. Unrelated relapses developed after a longer disease-free interval and tended to show poorer outcome compared with related relapse. In summary, relapse of a lymphoma from an unrelated clone is uncommon, but can occur in B-cell lymphomas. Clonal relationships should be determined by sequencing of the Ig genes, and not just by comparing the PCR product size.
Collapse
Affiliation(s)
- Seung Eun Lee
- Department of Pathology, Konkuk University School of Medicine, Konkuk University Medical Center, Seoul, Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hae Yong Yoo
- Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Seog Kim
- Division of Hematology and Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Hyeh Ko
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Mature lymphoid malignancies: origin, stem cells, and chronicity. Blood Adv 2017; 1:2444-2455. [PMID: 29296894 DOI: 10.1182/bloodadvances.2017008854] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/10/2017] [Indexed: 12/15/2022] Open
Abstract
The chronic behavior of mature lymphoid malignancies, with relapses occurring years apart in many patients, has until recently been unexplained. Patterns of relapse also differ vastly between disease entities, with some being highly curable by chemotherapy whereas others are destined to reemerge after treatment. Lately, the use of next-generation sequencing techniques has revealed essential information on the clonal evolution of lymphoid malignancies. Also, experimental xenograft transplantation point to the possible existence of an ancestral (stem) cell. Such a malignant lymphoid stem cell population could potentially evade current therapies and be the cause of chronicity and death in lymphoma patients; however, the evidence is divergent across disease entities and between studies. In this review we present an overview of genetic studies, case reports, and experimental evidence of the source of mature lymphoid malignancy and discuss the perspectives.
Collapse
|
31
|
Broséus J, Mourah S, Ramstein G, Bernard S, Mounier N, Cuccuini W, Gaulard P, Gisselbrecht C, Brière J, Houlgatte R, Thieblemont C. VEGF 121, is predictor for survival in activated B-cell-like diffuse large B-cell lymphoma and is related to an immune response gene signature conserved in cancers. Oncotarget 2017; 8:90808-90824. [PMID: 29207605 PMCID: PMC5710886 DOI: 10.18632/oncotarget.19385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/03/2017] [Indexed: 02/07/2023] Open
Abstract
Tumor microenvironment including endothelial and immune cells plays a crucial role in tumor progression and has been shown to dramatically influence cancer survival. In this study, we investigated the clinical relevance of the gene expression of key mediators of angiogenesis, VEGF isoforms 121, 165, and 189, and their receptors (VEGFR-1 and R-2) in a cohort of patients (n = 37) with relapsed/refractory diffuse large B-cell lymphoma (DLBCL) from the Collaborative Trial in Relapsed Aggressive Lymphoma (CORAL). In patients with ABC-like DLBCL, but not in patients with GCB-like DLBCL, low VEGF121 expression was associated with a significantly better survival than in those with high VEGF121 level: 4-year overall survival at 100% vs 36% (p = .011), respectively. A specific gene signature including 57 genes was correlated to VEGF121 expression level and was analyzed using a discovery process in 1,842 GSE datasets of public microarray studies. This gene signature was significantly expressed in other cancer datasets and was associated with immune response. In conclusion, low VEGF121 expression level was significantly associated with a good prognosis in relapsed/refractory ABC-like DLBCL, and with a well-conserved gene-expression profiling signature related to immune response. These findings pave the way for rationalization of drugs targeting immune response in refractory/relapsed ABC-like DLBCL.
Collapse
Affiliation(s)
- Julien Broséus
- Inserm U954, Faculty of Medicine, University of Lorraine, Nancy, France.,University Hospital of Nancy, Hematology Laboratory, Nancy, France
| | - Samia Mourah
- Paris Diderot University, Sorbonne Paris Cité, Paris, France.,APHP, Saint Louis University Hospital, Pharmacology-Biologic Laboratory, Paris, France.,Inserm UMRS 976, France
| | | | - Sophie Bernard
- APHP, Saint-Louis University Hospital, Hemato-Oncology, Paris, France
| | | | - Wendy Cuccuini
- APHP, Saint-Louis University Hospital, Hematology Laboratory, Paris, France
| | - Philippe Gaulard
- Department of Pathology, APHP, Henri Mondor University Hospital, Creteil, France.,Inserm U955, University Paris-Est, Créteil, France
| | - Christian Gisselbrecht
- APHP, Saint-Louis University Hospital, Hemato-Oncology, Paris, France.,Lymphoma Study Association, Pierre-Bénite, France
| | - Josette Brière
- Department of Pathology, APHP, Saint-Louis University Hospital, Paris, France
| | - Rémi Houlgatte
- Inserm U954, Faculty of Medicine, University of Lorraine, Nancy, France.,University Hospital of Nancy, DRCI, Nancy, France
| | - Catherine Thieblemont
- Paris Diderot University, Sorbonne Paris Cité, Paris, France.,APHP, Saint-Louis University Hospital, Hemato-Oncology, Paris, France
| |
Collapse
|
32
|
Juskevicius D, Dirnhofer S, Tzankov A. Genetic background and evolution of relapses in aggressive B-cell lymphomas. Haematologica 2017; 102:1139-1149. [PMID: 28554945 PMCID: PMC5566014 DOI: 10.3324/haematol.2016.151647] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022] Open
Abstract
Relapses of aggressive B-cell lymphomas pose a higher risk to affected patients because of potential treatment resistance and usually rapid tumor growth. Recent advances, such as targeting Bruton tyrosine kinase, have provided promising results in small numbers of cases, but treatment for the majority of patients remains challenging and outcomes are generally poor. A number of recent studies have utilized state-of-the-art genomic technologies in an attempt to better understand tumor genome evolution during relapse and to identify relapse-specific genetic alterations. It has been found that in some settings (e.g. diffuse large B-cell lymphomas in immunocompromised patients, secondary diffuse large B-cell lymphomas as Richter transformations) a significant part of the recurrences are clonally-unrelated de novo neoplasms, which might have distinct genomic and drug sensitivity profiles as well as different prognoses. Similar to earlier findings in indolent lymphomas, genetic tumor evolution of clonally-related relapsing aggressive B-cell lymphomas is predominantly characterized by two patterns: early divergence from a common progenitor and late divergence/linear evolution from a primary tumor. The clinical implications of these distinct patterns are not yet clear and will require additional investigation; however, it is plausible that these two patterns of recurrence are linked to different treatment-resistance mechanisms. Attempts to identify drivers of relapses result in a very heterogeneous list of affected genes and pathways as well as epigenetic mechanisms and suggest many ways of how recurrent tumors can adapt to treatment and expand their malignant properties.
Collapse
Affiliation(s)
- Darius Juskevicius
- Institute of Pathology & Medical Genetics, University of Basel and University Hospital Basel, Switzerland
| | - Stephan Dirnhofer
- Institute of Pathology & Medical Genetics, University of Basel and University Hospital Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Pathology & Medical Genetics, University of Basel and University Hospital Basel, Switzerland
| |
Collapse
|
33
|
Abstract
Diffuse large B-cell lymphoma (DLBCL) is divided into germinal center B-like (GCB) DLBCL and activated B-like (ABC) DLBCL. In recent years, whole genome sequencing (WGS), whole exome sequencing (WES), and transcriptome sequencing (RNA-seq) have been performed for samples from many patients with DLBCL. Here, I present a review of the results of next generation sequencing data for DLBCL. Somatic mutations show a low identity between studies with only 10-20% gene overlap. DLBCL is a disease that results from various molecular pathogeneses. Mutations in genes involved in chromatin remodeling were found in the GCB subtype. Mutations in members of B-cell receptor (BCR) signaling and the NF-κB pathway (MYD88) were found in the ABC subtype. The MYD88 L265P mutation was observed in 29% of ABC DLBCL cases. EZH2 mutations were observed in 21.7% of GCB DLBCL cases. WGS indicated that inactivating mutations in GNA13 (Gα protein) were prevalent in GCB DLBCL cases. In addition, S1PR2 is a target of aberrant somatic hypermutation. In recent years, samples from patients with relapsed and refractory DLBCL were analyzed. The activation of the NF-κB pathway is associated with treatment resistance in DLBCL. Further clarification of the molecular pathogenesis of DLBCL is expected to lead to the development of individualized treatment for the disease.
Collapse
Affiliation(s)
- Akito Dobashi
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research
| |
Collapse
|
34
|
A Survey of Computational Tools to Analyze and Interpret Whole Exome Sequencing Data. Int J Genomics 2016; 2016:7983236. [PMID: 28070503 PMCID: PMC5192301 DOI: 10.1155/2016/7983236] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 10/26/2016] [Indexed: 12/31/2022] Open
Abstract
Whole Exome Sequencing (WES) is the application of the next-generation technology to determine the variations in the exome and is becoming a standard approach in studying genetic variants in diseases. Understanding the exomes of individuals at single base resolution allows the identification of actionable mutations for disease treatment and management. WES technologies have shifted the bottleneck in experimental data production to computationally intensive informatics-based data analysis. Novel computational tools and methods have been developed to analyze and interpret WES data. Here, we review some of the current tools that are being used to analyze WES data. These tools range from the alignment of raw sequencing reads all the way to linking variants to actionable therapeutics. Strengths and weaknesses of each tool are discussed for the purpose of helping researchers make more informative decisions on selecting the best tools to analyze their WES data.
Collapse
|
35
|
Coiffier B, Sarkozy C. Diffuse large B-cell lymphoma: R-CHOP failure-what to do? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:366-378. [PMID: 27913503 PMCID: PMC6142522 DOI: 10.1182/asheducation-2016.1.366] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Although rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) is the standard treatment for patients with diffuse large B-cell lymphoma (DLBCL), ∼30% to 50% of patients are not cured by this treatment, depending on disease stage or prognostic index. Among patients for whom R-CHOP therapy fails, 20% suffer from primary refractory disease (progress during or right after treatment) whereas 30% relapse after achieving complete remission (CR). Currently, there is no good definition enabling us to identify these 2 groups upon diagnosis. Most of the refractory patients exhibit double-hit lymphoma (MYC-BCL2 rearrangement) or double-protein-expression lymphoma (MYC-BCL2 hyperexpression) which have a more aggressive clinical picture. New strategies are currently being explored to obtain better CR rates and fewer relapses. Although young relapsing patients are treated with high-dose therapy followed by autologous transplant, there is an unmet need for better salvage regimens in this setting. To prevent relapse, maintenance therapy with immunomodulatory agents such as lenalidomide is currently undergoing investigation. New drugs will most likely be introduced over the next few years and will probably be different for relapsing and refractory patients.
Collapse
Affiliation(s)
- Bertrand Coiffier
- Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Clémentine Sarkozy
- Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| |
Collapse
|
36
|
Choi S, Go JH, Kim EK, Lee H, Lee WM, Cho CS, Han K. Mutational Analysis of Extranodal NK/T-Cell Lymphoma Using Targeted Sequencing with a Comprehensive Cancer Panel. Genomics Inform 2016; 14:78-84. [PMID: 27729836 PMCID: PMC5056900 DOI: 10.5808/gi.2016.14.3.78] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 02/06/2023] Open
Abstract
Extranodal natural killer (NK)/T-cell lymphoma, nasal type (NKTCL), is a malignant disorder of cytotoxic lymphocytes of NK or T cells. It is an aggressive neoplasm with a very poor prognosis. Although extranodal NKTCL reportedly has a strong association with Epstein-Barr virus, the molecular pathogenesis of NKTCL has been unexplored. The recent technological advancements in next-generation sequencing (NGS) have made DNA sequencing cost- and time-effective, with more reliable results. Using the Ion Proton Comprehensive Cancer Panel, we sequenced 409 cancer-related genes to identify somatic mutations in five NKTCL tissue samples. The sequencing analysis detected 25 mutations in 21 genes. Among them, KMT2D, a histone modification-related gene, was the most frequently mutated gene (four of the five cases). This result was consistent with recent NGS studies that have suggested KMT2D as a novel driver gene in NKTCL. Mutations were also found in ARID1A, a chromatin remodeling gene, and TP53, which also recurred in recent NGS studies. We also found mutations in 18 novel candidate genes, with molecular functions that were potentially implicated in cancer development. We suggest that these genes may result in multiple oncogenic events and may be used as potential bio-markers of NKTCL in the future.
Collapse
Affiliation(s)
- Seungkyu Choi
- Department of Pathology, Dankook University College of Medicine, Cheonan 31116, Korea
| | - Jai Hyang Go
- Department of Pathology, Dankook University College of Medicine, Cheonan 31116, Korea
| | - Eun Kyung Kim
- Department of Pathology, Eulji Medical Center, Eulji University School of Medicine, Seoul 01830, Korea
| | - Hojung Lee
- Department of Pathology, Eulji Medical Center, Eulji University School of Medicine, Seoul 01830, Korea
| | - Won Mi Lee
- Department of Pathology, Eulji Medical Center, Eulji University School of Medicine, Seoul 01830, Korea
| | - Chun-Sung Cho
- Department of Neurosurgery, Dankook University College of Medicine, Cheonan 31116, Korea
| | - Kyudong Han
- Department of Nanobiomedical Science, BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea
| |
Collapse
|
37
|
Distinct genetic evolution patterns of relapsing diffuse large B-cell lymphoma revealed by genome-wide copy number aberration and targeted sequencing analysis. Leukemia 2016; 30:2385-2395. [PMID: 27198204 DOI: 10.1038/leu.2016.135] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/29/2016] [Accepted: 05/03/2016] [Indexed: 01/16/2023]
Abstract
Recurrences of diffuse large B-cell lymphomas (DLBCL) result in significant morbidity and mortality, but their underlying genetic and biological mechanisms are unclear. Clonal relationship in DLBCL relapses so far is mostly addressed by the investigation of immunoglobulin (IG) rearrangements, therefore, lacking deeper insights into genome-wide lymphoma evolution. We studied mutations and copy number aberrations in 20 paired relapsing and 20 non-relapsing DLBCL cases aiming to test the clonal relationship between primaries and relapses to track tumors' genetic evolution and to investigate the genetic background of DLBCL recurrence. Three clonally unrelated DLBCL relapses were identified (15%). Also, two distinct patterns of genetic evolution in clonally related relapses were detected as follows: (1) early-divergent/branching evolution from a common progenitor in 6 patients (30%), and (2) late-divergent/linear progression of relapses in 11 patients (65%). Analysis of recurrent genetic events identified potential early drivers of lymphomagenesis (KMT2D, MYD88, CD79B and PIM1). The most frequent relapse-specific events were additional mutations in KMT2D and alterations of MEF2B. SOCS1 mutations were exclusive to non-relapsing DLBCL, whereas primaries of relapsing DLBCL more commonly displayed gains of 10p15.3-p12.1 containing the potential oncogenes PRKCQ, GATA3, MLLT10 and ABI1. Altogether, our study expands the knowledge on clonal relationship, genetic evolution and mutational basis of DLBCL relapses.
Collapse
|
38
|
Phase 2 study of panobinostat with or without rituximab in relapsed diffuse large B-cell lymphoma. Blood 2016; 128:185-94. [PMID: 27166360 DOI: 10.1182/blood-2016-02-699520] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/22/2016] [Indexed: 12/16/2022] Open
Abstract
The majority of diffuse large B-cell lymphoma (DLBCL) tumors contain mutations in histone-modifying enzymes (HMEs), indicating a potential therapeutic benefit of histone deacetylase inhibitors (HDIs), and preclinical data suggest that HDIs augment the effect of rituximab. In this randomized phase 2 study, we evaluated the response rate and toxicity of panobinostat, a pan-HDI administered 30 mg orally 3 times weekly, with or without rituximab, in 40 patients with relapsed or refractory de novo (n = 27) or transformed (n = 13) DLBCL. Candidate genes and whole exomes were sequenced in relapse tumor biopsies to search for molecular correlates, and these data were used to quantify circulating tumor DNA (ctDNA) in serial plasma samples. Eleven of 40 patients (28%) responded to panobinostat (95% confidence interval [CI] 14.6-43.9) and rituximab did not increase responses. The median duration of response was 14.5 months (95% CI 9.4 to "not reached"). At time of data censoring, 6 of 11 patients had not progressed. Of the genes tested for mutations, only those in MEF2B were significantly associated with response. We detected ctDNA in at least 1 plasma sample from 96% of tested patients. A significant increase in ctDNA at day 15 relative to baseline was strongly associated with lack of response (sensitivity 71.4%, specificity 100%). We conclude that panobinostat induces very durable responses in some patients with relapsed DLBCL, and early responses can be predicted by mutations in MEF2B or a significant change in ctDNA level at 15 days after treatment initiation. This clinical trial was registered at www.ClinicalTrials.gov (#NCT01238692).
Collapse
|
39
|
Dubois S, Mareschal S, Picquenot JM, Viailly PJ, Bohers E, Cornic M, Bertrand P, Veresezan EL, Ruminy P, Maingonnat C, Marchand V, Lanic H, Penther D, Bastard C, Tilly H, Jardin F. Immunohistochemical and genomic profiles of diffuse large B-cell lymphomas: implications for targeted EZH2 inhibitor therapy? Oncotarget 2016; 6:16712-24. [PMID: 25762637 PMCID: PMC4599301 DOI: 10.18632/oncotarget.3154] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/15/2015] [Indexed: 11/25/2022] Open
Abstract
Enhancer of Zeste Homolog 2 (EZH2) plays an essential epigenetic role in Diffuse Large B Cell Lymphoma (DLBCL) development. Recurrent somatic heterozygous gain-of-function mutations of EZH2 have been identified in DLBCL, most notably affecting tyrosine 641 (Y641), inducing hyper-trimethylation of H3K27 (H3K27me3). Novel EZH2 inhibitors are being tested in phase 1 and 2 clinical trials but no study has examined which patients would most benefit from this treatment. We evaluated the immunohistochemical (IHC) methylation profiles of 82 patients with DLBCL, as well as the mutational profiles of 32 patients with DLBCL using NGS analysis of a panel of 34 genes involved in lymphomagenesis. A novel IHC score based on H3K27me2 and H3K27me3 expression was developed, capable of distinguishing patients with wild-type (WT) EZH2 and patients with EZH2 Y641 mutations (p = 10−5). NGS analysis revealed a subclonal EZH2 mutation pattern in EZH2 mutant patients with WT-like IHC methylation profiles, while associated mutations capable of upregulating EZH2 were detected in WT EZH2 patients with mutant-like IHC methylation profiles. IHC and mutational profiles highlight in vivo hyper-H3K27me3 and hypo-H3K27me2 status, pinpoint associated activating mutations and determine EZH2 mutation clonality, maximizing EZH2 inhibitor potential by identifying patients most likely to benefit from treatment.
Collapse
Affiliation(s)
- Sydney Dubois
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Sylvain Mareschal
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Jean-Michel Picquenot
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.,Department of Pathology, Centre Henri Becquerel, Rouen, France
| | | | - Elodie Bohers
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Marie Cornic
- Department of Pathology, Centre Henri Becquerel, Rouen, France
| | - Philippe Bertrand
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Elena Liana Veresezan
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.,Department of Pathology, Centre Henri Becquerel, Rouen, France
| | - Philippe Ruminy
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | | | - Vinciane Marchand
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Hélène Lanic
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.,Department of Clinical Hematology, Centre Henri Becquerel, Rouen, France
| | - Dominique Penther
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Christian Bastard
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France
| | - Hervé Tilly
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.,Department of Clinical Hematology, Centre Henri Becquerel, Rouen, France
| | - Fabrice Jardin
- INSERM U918, Centre Henri Becquerel, Université de Rouen, IRIB, Rouen, France.,Department of Clinical Hematology, Centre Henri Becquerel, Rouen, France
| |
Collapse
|
40
|
Jiang Y, Nie K, Redmond D, Melnick AM, Tam W, Elemento O. VDJ-Seq: Deep Sequencing Analysis of Rearranged Immunoglobulin Heavy Chain Gene to Reveal Clonal Evolution Patterns of B Cell Lymphoma. J Vis Exp 2015:e53215. [PMID: 26780364 DOI: 10.3791/53215] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Understanding tumor clonality is critical to understanding the mechanisms involved in tumorigenesis and disease progression. In addition, understanding the clonal composition changes that occur within a tumor in response to certain micro-environment or treatments may lead to the design of more sophisticated and effective approaches to eradicate tumor cells. However, tracking tumor clonal sub-populations has been challenging due to the lack of distinguishable markers. To address this problem, a VDJ-seq protocol was created to trace the clonal evolution patterns of diffuse large B cell lymphoma (DLBCL) relapse by exploiting VDJ recombination and somatic hypermutation (SHM), two unique features of B cell lymphomas. In this protocol, Next-Generation sequencing (NGS) libraries with indexing potential were constructed from amplified rearranged immunoglobulin heavy chain (IgH) VDJ region from pairs of primary diagnosis and relapse DLBCL samples. On average more than half million VDJ sequences per sample were obtained after sequencing, which contain both VDJ rearrangement and SHM information. In addition, customized bioinformatics pipelines were developed to fully utilize sequence information for the characterization of IgH-VDJ repertoire within these samples. Furthermore, the pipeline allows the reconstruction and comparison of the clonal architecture of individual tumors, which enables the examination of the clonal heterogeneity within the diagnosis tumors and deduction of clonal evolution patterns between diagnosis and relapse tumor pairs. When applying this analysis to several diagnosis-relapse pairs, we uncovered key evidence that multiple distinctive tumor evolutionary patterns could lead to DLBCL relapse. Additionally, this approach can be expanded into other clinical aspects, such as identification of minimal residual disease, monitoring relapse progress and treatment response, and investigation of immune repertoires in non-lymphoma contexts.
Collapse
Affiliation(s)
- Yanwen Jiang
- Department of Medicine, Weill Cornell Medical College; Institute for Computational Biomedicine, Weill Cornell Medical College;
| | - Kui Nie
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College
| | - David Redmond
- Institute for Computational Biomedicine, Weill Cornell Medical College
| | - Ari M Melnick
- Department of Medicine, Weill Cornell Medical College
| | - Wayne Tam
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College;
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medical College;
| |
Collapse
|
41
|
|
42
|
Apostoli AJ, Ailles L. Clonal evolution and tumor-initiating cells: New dimensions in cancer patient treatment. Crit Rev Clin Lab Sci 2015; 53:40-51. [PMID: 26397062 DOI: 10.3109/10408363.2015.1083944] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human cancer is not a uniform disease but a plethora of disparate tumor types and subtypes. The differences that exist between individual tumors (intertumoral heterogeneity) present a significant roadblock to the eradication of cancer. It has also become increasingly clear that variations across individual tumors (intratumoral heterogeneity) have important implications to cancer progression and treatment efficacy. Therefore, in order to improve patient care and develop novel chemotherapeutics, the evolving tumor landscape needs to be further explored. Next-generation sequencing (NGS) technologies are revolutionizing the cancer research arena by providing state-of-the-art, high-speed methods of genome sequencing at single-nucleotide resolution, thus enabling an unprecedented detection of tumor-specific genetic abnormalities. These anomalies can be quantified to reveal specific frequencies of DNA alterations that correspond to distinct clonal populations within a given tumor. As such, NGS approaches have also been utilized to explore the heterogeneous landscape of patient tumors as well as to match metastatic and/or recurrent growths and patient-derived engrafts. By sequencing in this manner--through time so to speak--cancer researchers can track shifting clonal populations, make important inferences about tumor evolution and potentially identify tumor subclones that could be viably targeted. This exciting new territory has important implications for the competing clonal evolution and cancer stem cell models of tumor heterogeneity, and also offers a new dimension for cancer treatment and profound hope for patients in the coming years.
Collapse
Affiliation(s)
- Anthony J Apostoli
- a Princess Margaret Cancer Centre, University Health Network , Toronto , Ontario , Canada and
| | - Laurie Ailles
- a Princess Margaret Cancer Centre, University Health Network , Toronto , Ontario , Canada and.,b Department of Medical Biophysics , University of Toronto , Toronto , Ontario , Canada
| |
Collapse
|
43
|
Carlotti E, Wrench D, Rosignoli G, Marzec J, Sangaralingam A, Hazanov L, Michaeli M, Hallam S, Chaplin T, Iqbal S, Calaminici M, Young B, Mehr R, Campbell P, Fitzgibbon J, Gribben JG. High Throughput Sequencing Analysis of the Immunoglobulin Heavy Chain Gene from Flow-Sorted B Cell Sub-Populations Define the Dynamics of Follicular Lymphoma Clonal Evolution. PLoS One 2015; 10:e0134833. [PMID: 26325507 PMCID: PMC4556522 DOI: 10.1371/journal.pone.0134833] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 07/14/2015] [Indexed: 11/19/2022] Open
Abstract
Understanding the dynamics of evolution of Follicular Lymphoma (FL) clones during disease progression is important for monitoring and targeting this tumor effectively. Genetic profiling of serial FL biopsies and examples of FL transmission following bone marrow transplant suggest that this disease may evolve by divergent evolution from a common ancestor cell. However where this ancestor cell resides and how it evolves is still unclear. The analysis of the pattern of somatic hypermutation of the immunoglobulin gene (Ig) is traditionally used for tracking the physiological clonal evolution of B cells within the germinal center and allows to discriminate those cells that have just entered the germinal center and display features of ancestor cells from those B cells that keep re-circulating across different lymphoid organs. Here we investigated the pattern of somatic hypermutation of the heavy chain of the immunoglobulin gene (IgH-VH) in 4 flow-sorted B cells subpopulations belonging to different stages of differentiation, from sequential lymph node biopsies of cases displaying diverse patterns of evolution, using the GS-FLX Titanium sequencing platform. We observed an unexpectedly high level of clonality, with hundreds of distinct tumor subclones in the different subpopulations from the same sample, the majority detected at a frequency <10-2. By using a lineage trees analysis we observed in all our FL and t-FL cases that the oligoclonal FL population was trapped in a narrow intermediate stage of maturation that maintains the capacity to undergo SHM, but was unable to further differentiate. The presence of such a complex architecture highlights challenges currently encountered in finding a cure for this disease.
Collapse
Affiliation(s)
- Emanuela Carlotti
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - David Wrench
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Guglielmo Rosignoli
- Flow Cytometry Core Facility, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Jacek Marzec
- Centre for Molecular Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Ajanthah Sangaralingam
- Centre for Molecular Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Lena Hazanov
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Miri Michaeli
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Simon Hallam
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Tracy Chaplin
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Sameena Iqbal
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Maria Calaminici
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Bryan Young
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - Ramit Mehr
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Peter Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Jude Fitzgibbon
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| | - John G. Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute - a CR-UK Centre Of Excellence, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
44
|
Miles G, Rae J, Ramalingam SS, Pfeifer J. Genetic Testing and Tissue Banking for Personalized Oncology: Analytical and Institutional Factors. Semin Oncol 2015; 42:713-23. [PMID: 26433552 DOI: 10.1053/j.seminoncol.2015.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Personalized oncology, or more aptly precision oncogenomics, refers to the identification and implementation of clinically actionable targets tailored to an individual patient's cancer genomic information. Banking of human tissue and other biospecimens establishes a framework to extract and collect the data essential to our understanding of disease pathogenesis and treatment. Cancer cooperative groups in the United States have led the way in establishing robust biospecimen collection mechanisms to facilitate translational research, and combined with technological advances in molecular testing, tissue banking has expanded from its traditional base in academic research and is assuming an increasingly pivotal role in directing the clinical care of cancer patients. Comprehensive screening of tumors by DNA sequencing and the ability to mine and interpret these large data sets from well-organized tissue banks have defined molecular subtypes of cancer. Such stratification by genomic criteria has revolutionized our perspectives on cancer diagnosis and treatment, offering insight into prognosis, progression, and susceptibility or resistance to known therapeutic agents. In turn, this has enabled clinicians to offer treatments tailored to patients that can greatly improve their chances of survival. Unique challenges and opportunities accompany the rapidly evolving interplay between tissue banking and genomic sequencing, and are the driving forces underlying the revolution in precision medicine. Molecular testing and precision medicine clinical trials are now becoming the major thrust behind the cooperative groups' clinical research efforts.
Collapse
Affiliation(s)
- George Miles
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.
| | - James Rae
- Department of Internal Medicine & Pharmacology, University of Michigan, Ann Arbor, MI
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory School of Medicine, Winship Cancer Institute, Atlanta, GA
| | - John Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
45
|
Epigenomic evolution in diffuse large B-cell lymphomas. Nat Commun 2015; 6:6921. [PMID: 25891015 PMCID: PMC4411286 DOI: 10.1038/ncomms7921] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 03/16/2015] [Indexed: 12/24/2022] Open
Abstract
The contribution of epigenomic alterations to tumour progression and relapse is not well characterized. Here we characterize an association between disease progression and DNA methylation in diffuse large B-cell lymphoma (DLBCL). By profiling genome-wide DNA methylation at single-base pair resolution in thirteen DLBCL diagnosis–relapse sample pairs, we show that DLBCL patients exhibit heterogeneous evolution of tumour methylomes during relapse. We identify differentially methylated regulatory elements and determine a relapse-associated methylation signature converging on key pathways such as transforming growth factor-β (TGF-β) receptor activity. We also observe decreased intra-tumour methylation heterogeneity from diagnosis to relapsed tumour samples. Relapse-free patients display lower intra-tumour methylation heterogeneity at diagnosis compared with relapsed patients in an independent validation cohort. Furthermore, intra-tumour methylation heterogeneity is predictive of time to relapse. Therefore, we propose that epigenomic heterogeneity may support or drive the relapse phenotype and can be used to predict DLBCL relapse. The contribution of epigenomic alterations to tumour progression and relapse is not well characterized. Here the authors characterize epigenetic evolution in aggressive B-cell lymphoma and find that epigenomic heterogeneity may not only support and drive the relapse phenotype but also be used to predict lymphoma relapse.
Collapse
|
46
|
Abstract
The pathogenesis of diffuse large B-cell lymphoma (DLBCL) is strongly linked to perturbation of epigenetic mechanisms. The germinal center (GC) B cells from which DLBCLs arise are prone to instability in their cytosine methylation patterns. DLBCLs inherit this epigenetic instability and display variable degrees of epigenetic heterogeneity. Greater epigenetic heterogeneity is linked with poor clinical outcome. Somatic mutations of histone-modifying proteins have also emerged as a hallmark of DLBCL. The effect of these somatic mutations may be to disrupt epigenetic switches that control the GC phenotype and "lock in" certain oncogenic features of GC B cells, resulting in malignant transformation. DNA methyltransferase and histone methyltransferase inhibitors are emerging as viable therapeutic approaches to erase aberrant epigenetic programming, suppress DLBCL growth, and overcome chemotherapy resistance. This review will discuss these recent advances and their therapeutic implications.
Collapse
Affiliation(s)
- Yanwen Jiang
- Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Ari Melnick
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|