151
|
Chen L, Pan X, Hu X, Zhang YH, Wang S, Huang T, Cai YD. Gene expression differences among different MSI statuses in colorectal cancer. Int J Cancer 2018; 143:1731-1740. [PMID: 29696646 DOI: 10.1002/ijc.31554] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/30/2018] [Accepted: 04/18/2018] [Indexed: 12/28/2022]
Abstract
Colorectal cancer is the third most common cancer in males and second in females. This disease can be caused by genetic and acquired/environmental factors. Microsatellite instability (MSI) is one of the major mechanisms in colorectal cancer. This mechanism is a specific condition of genetic hyper mutability that results from incompetent DNA mismatch repair. MSI has been applied to classify different colorectal cancer subtypes. However, the effects of MSI status on gene expression are largely unknown. In our study, we integrated the gene expression profile and MSI status of all CRC samples from the TCGA database, and then categorized the CRC samples into three subgroups, namely, MSI-stable, MSI-low, and MSI-high, according to the MSI status. We applied a novel computational method based on machine learning and screened the genes specifically expressed for the different colorectal cancer subtypes. The results showed the distinct mechanisms of the different colorectal cancer subtypes with MSI status and provided the genes that may be the optimal standards to further classify the various molecular subtypes of colorectal cancer with distinct MSI status.
Collapse
Affiliation(s)
- Lei Chen
- College of Life Science, Shanghai University, Shanghai, 200444, People' Republic of China.,College of Information Engineering, Shanghai Maritime University, Shanghai, 201306, People's Republic of China
| | - Xiaoyong Pan
- Department of Medical Informatics, Erasmus MC, Rotterdam, Netherlands
| | - XiaoHua Hu
- Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - ShaoPeng Wang
- College of Life Science, Shanghai University, Shanghai, 200444, People' Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, People's Republic of China
| | - Yu-Dong Cai
- College of Life Science, Shanghai University, Shanghai, 200444, People' Republic of China
| |
Collapse
|
152
|
Hempelmann JA, Lockwood CM, Konnick EQ, Schweizer MT, Antonarakis ES, Lotan TL, Montgomery B, Nelson PS, Klemfuss N, Salipante SJ, Pritchard CC. Microsatellite instability in prostate cancer by PCR or next-generation sequencing. J Immunother Cancer 2018; 6:29. [PMID: 29665853 PMCID: PMC5904988 DOI: 10.1186/s40425-018-0341-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/04/2018] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Microsatellite instability (MSI) is now being used as a sole biomarker to guide immunotherapy treatment for men with advanced prostate cancer. Yet current molecular diagnostic tests for MSI have not been evaluated for use in prostate cancer. METHODS We evaluated two next-generation sequencing (NGS) MSI-detection methods, MSIplus (18 markers) and MSI by Large Panel NGS (> 60 markers), and compared the performance of each NGS method to the most widely used 5-marker MSI-PCR detection system. All methods were evaluated by comparison to targeted whole gene sequencing of DNA mismatch-repair genes, and immunohistochemistry for mismatch repair genes, where available. RESULTS In a set of 91 prostate tumors with known mismatch repair status (29-deficient and 62-intact mismatch-repair) MSIplus had a sensitivity of 96.6% (28/29) and a specificity of 100% (62/62), MSI by Large Panel NGS had a sensitivity of 93.1% (27/29) and a specificity of 98.4% (61/62), and MSI-PCR had a sensitivity of 72.4% (21/29) and a specificity of 100% (62/62). CONCLUSIONS We found that the widely used 5-marker MSI-PCR panel has inferior sensitivity when applied to prostate cancer and that NGS testing with an expanded panel of markers performs well. In addition, NGS methods offer advantages over MSI-PCR, including no requirement for matched non-tumor tissue and an automated analysis pipeline with quantitative interpretation of MSI-status.
Collapse
Affiliation(s)
| | | | - Eric Q Konnick
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Michael T Schweizer
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Emmanuel S Antonarakis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bruce Montgomery
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
- VA Puget Sound Health Care System, Seattle, WA, USA
| | - Peter S Nelson
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nola Klemfuss
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Stephen J Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
153
|
Gray PN, Tsai P, Chen D, Wu S, Hoo J, Mu W, Li B, Vuong H, Lu HM, Batth N, Willett S, Uyeda L, Shah S, Gau CL, Umali M, Espenschied C, Janicek M, Brown S, Margileth D, Dobrea L, Wagman L, Rana H, Hall MJ, Ross T, Terdiman J, Cullinane C, Ries S, Totten E, Elliott AM. TumorNext-Lynch-MMR: a comprehensive next generation sequencing assay for the detection of germline and somatic mutations in genes associated with mismatch repair deficiency and Lynch syndrome. Oncotarget 2018; 9:20304-20322. [PMID: 29755653 PMCID: PMC5945525 DOI: 10.18632/oncotarget.24854] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022] Open
Abstract
The current algorithm for Lynch syndrome diagnosis is highly complex with multiple steps which can result in an extended time to diagnosis while depleting precious tumor specimens. Here we describe the analytical validation of a custom probe-based NGS tumor panel, TumorNext-Lynch-MMR, which generates a comprehensive genetic profile of both germline and somatic mutations that can accelerate and streamline the time to diagnosis and preserve specimen. TumorNext-Lynch-MMR can detect single nucleotide variants, small insertions and deletions in 39 genes that are frequently mutated in Lynch syndrome and colorectal cancer. Moreover, the panel provides microsatellite instability status and detects loss of heterozygosity in the five Lynch genes; MSH2, MSH6, MLH1, PMS2 and EPCAM. Clinical cases are described that highlight the assays ability to differentiate between somatic and germline mutations, precisely classify variants and resolve discordant cases.
Collapse
Affiliation(s)
- Phillip N Gray
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Pei Tsai
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Daniel Chen
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Sitao Wu
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Jayne Hoo
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Wenbo Mu
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Bing Li
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Huy Vuong
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Hsiao-Mei Lu
- Bioinformatics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Navanjot Batth
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Sara Willett
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Lisa Uyeda
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Swati Shah
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Chia-Ling Gau
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Monalyn Umali
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Carin Espenschied
- Clinical Diagnostics Department, Ambry Genetics, Aliso Viejo, CA 92656, USA
| | - Mike Janicek
- Cancer Genetic Risk Assessment Program, Arizona Oncology, Scottsdale, AZ 85258, USA
| | - Sandra Brown
- Cancer Genetics Program, Saint Joseph of Orange, Orange, CA 92868, USA
| | - David Margileth
- Cancer Genetics Program, Saint Joseph of Orange, Orange, CA 92868, USA
| | - Lavinia Dobrea
- Oncology Research and Biospecimen Program, Saint Joseph of Orange, Orange, CA 92868, USA
| | - Lawrence Wagman
- The Center for Cancer Prevention and Treatment, Saint Joseph of Orange, Orange, CA 92868, USA
| | - Huma Rana
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02461, USA
| | - Michael J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia PA 19111, USA
| | - Theodora Ross
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan Terdiman
- Department of Medicine - Gastroenterology, University of California San Francisco, San Francisco, CA 94115, USA
| | - Carey Cullinane
- Department of Pathology, Long Beach Memorial Medical Center, Long Beach, CA 90801, USA
| | - Savita Ries
- Department of Pathology, Long Beach Memorial Medical Center, Long Beach, CA 90801, USA
| | - Ellen Totten
- Advocate Medical Group, Park Ridge, Illinois 60068, USA
| | - Aaron M Elliott
- Advanced Genomic Services, Ambry Genetics, Aliso Viejo, CA 92656, USA
| |
Collapse
|
154
|
Abstract
Microsatellite instability (MSI) refers to the hypermutator phenotype secondary to frequent polymorphism in short repetitive DNA sequences and single nucleotide substitution, as consequence of DNA mismatch repair (MMR) deficiency. MSI secondary to germline mutation in DNA MMR proteins is the molecular fingerprint of Lynch syndrome (LS), while epigenetic inactivation of these genes is more commonly found in sporadic MSI tumors. MSI occurs at different frequencies across malignancies, although original methods to assess MSI or MMR deficiency have been developed mostly in LS related cancers. Here we will discuss the current methods to detect MSI/MMR deficiency with a focus of new tools which are emerging as highly sensitive detector for MSI across multiple tumor types. Due to high frequencies of non-synonymous mutations, the presence of frameshift-mutated neoantigens, which can trigger a more robust and long-lasting immune response and strong TIL infiltration with tumor eradication, MSI has emerged as an important predictor of sensitivity for immunotherapy-based strategies, as showed by the recent FDA's first histology agnostic-accelerated approval to immune checkpoint inhibitors for refractory, adult and pediatric, MMR deficient (dMMR) or MSI high (MSI-H) tumors. Moreover, it is known that MSI status may predict cancer response/resistance to certain chemotherapies. Here we will describe the complex interplay between the genetic and clinical-pathological features of MSI/dMMR tumors and the cancer immunotherapy, with a focus on the predictive and prognostic role of MMR status for immune checkpoint inhibitors (ICIs) and providing some suggestions on how to conceive better predictive markers for immunotherapy in the next future.
Collapse
Affiliation(s)
- Marina Baretti
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Hospital, United States
| | - Dung T Le
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins Hospital, United States.
| |
Collapse
|
155
|
Testa U, Pelosi E, Castelli G. Colorectal cancer: genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med Sci (Basel) 2018; 6:E31. [PMID: 29652830 PMCID: PMC6024750 DOI: 10.3390/medsci6020031] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/24/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023] Open
Abstract
Colon cancer is the third most common cancer worldwide. Most colorectal cancer occurrences are sporadic, not related to genetic predisposition or family history; however, 20-30% of patients with colorectal cancer have a family history of colorectal cancer and 5% of these tumors arise in the setting of a Mendelian inheritance syndrome. In many patients, the development of a colorectal cancer is preceded by a benign neoplastic lesion: either an adenomatous polyp or a serrated polyp. Studies carried out in the last years have characterized the main molecular alterations occurring in colorectal cancers, showing that the tumor of each patient displays from two to eight driver mutations. The ensemble of molecular studies, including gene expression studies, has led to two proposed classifications of colorectal cancers, with the identification of four/five non-overlapping groups. The homeostasis of the rapidly renewing intestinal epithelium is ensured by few stem cells present at the level of the base of intestinal crypts. Various experimental evidence suggests that colorectal cancers may derive from the malignant transformation of intestinal stem cells or of intestinal cells that acquire stem cell properties following malignant transformation. Colon cancer stem cells seem to be involved in tumor chemoresistance, radioresistance and relapse.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
156
|
Bailey MH, Tokheim C, Porta-Pardo E, Sengupta S, Bertrand D, Weerasinghe A, Colaprico A, Wendl MC, Kim J, Reardon B, Ng PKS, Jeong KJ, Cao S, Wang Z, Gao J, Gao Q, Wang F, Liu EM, Mularoni L, Rubio-Perez C, Nagarajan N, Cortés-Ciriano I, Zhou DC, Liang WW, Hess JM, Yellapantula VD, Tamborero D, Gonzalez-Perez A, Suphavilai C, Ko JY, Khurana E, Park PJ, Van Allen EM, Liang H, Lawrence MS, Godzik A, Lopez-Bigas N, Stuart J, Wheeler D, Getz G, Chen K, Lazar AJ, Mills GB, Karchin R, Ding L. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 2018; 173:371-385.e18. [PMID: 29625053 PMCID: PMC6029450 DOI: 10.1016/j.cell.2018.02.060] [Citation(s) in RCA: 1235] [Impact Index Per Article: 205.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/22/2017] [Accepted: 02/23/2018] [Indexed: 12/19/2022]
Abstract
Identifying molecular cancer drivers is critical for precision oncology. Multiple advanced algorithms to identify drivers now exist, but systematic attempts to combine and optimize them on large datasets are few. We report a PanCancer and PanSoftware analysis spanning 9,423 tumor exomes (comprising all 33 of The Cancer Genome Atlas projects) and using 26 computational tools to catalog driver genes and mutations. We identify 299 driver genes with implications regarding their anatomical sites and cancer/cell types. Sequence- and structure-based analyses identified >3,400 putative missense driver mutations supported by multiple lines of evidence. Experimental validation confirmed 60%-85% of predicted mutations as likely drivers. We found that >300 MSI tumors are associated with high PD-1/PD-L1, and 57% of tumors analyzed harbor putative clinically actionable events. Our study represents the most comprehensive discovery of cancer genes and mutations to date and will serve as a blueprint for future biological and clinical endeavors.
Collapse
Affiliation(s)
- Matthew H Bailey
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Collin Tokheim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Eduard Porta-Pardo
- Barcelona Supercomputing Centre (BSC), Barcelona, Spain; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sohini Sengupta
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Denis Bertrand
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 138672
| | - Amila Weerasinghe
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Antonio Colaprico
- Interuniversity Institute of Bioinformatics in Brussels (IB2), 1050 Brussels, Belgium; Machine Learning Group (MLG), Département d'Informatique, Université Libre de Bruxelles (ULB), Boulevard du Triomphe, CP212, 1050 Bruxelles, Belgium; Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Michael C Wendl
- McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA; Department of Mathematics, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jaegil Kim
- The Broad Institute, Cambridge, MA 02142, USA
| | - Brendan Reardon
- The Broad Institute, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Patrick Kwok-Shing Ng
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kang Jin Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Song Cao
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Zixing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjiong Gao
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qingsong Gao
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Fang Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Eric Minwei Liu
- Meyer Cancer Center and Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Loris Mularoni
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Carlota Rubio-Perez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Niranjan Nagarajan
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 138672
| | - Isidro Cortés-Ciriano
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Boston, MA 02115, USA; Centre for Molecular Science Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Daniel Cui Zhou
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - Wen-Wei Liang
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | | | - Venkata D Yellapantula
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA
| | - David Tamborero
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Abel Gonzalez-Perez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain
| | - Chayaporn Suphavilai
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 138672
| | - Jia Yu Ko
- Computational and Systems Biology, Genome Institute of Singapore, Singapore, 138672
| | - Ekta Khurana
- Meyer Cancer Center and Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Boston, MA 02115, USA
| | - Eliezer M Van Allen
- The Broad Institute, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Han Liang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael S Lawrence
- The Broad Institute, Cambridge, MA 02142, USA; Department of Pathology, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA 02114, USA
| | - Adam Godzik
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Nuria Lopez-Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Josh Stuart
- University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - David Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gad Getz
- The Broad Institute, Cambridge, MA 02142, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexander J Lazar
- Departments of Pathology, Genomic Medicine, & Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rachel Karchin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD 21287, USA.
| | - Li Ding
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University, St. Louis, MO 63108, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
157
|
Berger NA. Young Adult Cancer: Influence of the Obesity Pandemic. Obesity (Silver Spring) 2018; 26:641-650. [PMID: 29570247 PMCID: PMC5868416 DOI: 10.1002/oby.22137] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/07/2017] [Accepted: 01/11/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The purpose of this article is to review the association of the obesity pandemic with appearance of cancers in young adults under age 50 and to define potential mechanisms by which obesity may accelerate the development of malignancy. METHODS A comprehensive narrative review was performed to integrate preclinical, clinical, and epidemiologic evidence describing the association of obesity with cancer in young adults based on a search of PubMed and Google databases. RESULTS Results from more than 100 publications are summarized. Although they differ in age groups analyzed and incidence of obesity, sufficient data exists to suggest an influence of the obesity pandemic on the increase of cancer among young adults. CONCLUSIONS Cancer in young adults is occurring with increasing frequency. Overweight and obesity have become major public health issues reaching pandemic proportions. Excess weight is associated with increased cancer risk, morbidity, and mortality. Multiple murine models indicate that obesity not only increases cancer incidence but also accelerates its development. Thus, the possibility exists that overweight and obesity may be contributing to the appearance of specific malignancies at younger ages. This prospect, in association with the worldwide expansion of obesity, suggests an impending explosive increase in obesity-associated cancers in young adults.
Collapse
Affiliation(s)
- Nathan A Berger
- Hematology/Oncology Division, Departments of Medicine, Biochemistry, Genetics & Genome Sciences, Center for Science, Health, and Society, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
158
|
De Angelis ML, Bruselles A, Francescangeli F, Pucilli F, Vitale S, Zeuner A, Tartaglia M, Baiocchi M. Colorectal cancer spheroid biobanks: multi-level approaches to drug sensitivity studies. Cell Biol Toxicol 2018; 34:459-469. [PMID: 29478126 DOI: 10.1007/s10565-018-9423-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 02/06/2018] [Indexed: 12/30/2022]
Abstract
Biobanking of molecularly characterized colorectal cancer stem cells (CSCs) generated from individual patients and growing as spheroids in defined serum-free media offer a fast, feasible, and multi-level approach for the screening of targeted therapies and drug resistance molecular studies. By combining in vitro and in vivo analyses of cetuximab efficacy with genetic data on an ongoing collection of stem cell-enriched spheroids, we describe the identification and preliminary characterization of microsatellite stable (MSS) CSCs that, despite the presence of the KRAS (G12D) mutation, display epidermal growth factor (EGF)-dependent growth and are strongly inhibited by anti-EGF-receptor (EGFR) treatment. In parallel, we detected an increased resistance to anti-EGFR therapy of microsatellite instable (MSI) CSC lines irrespective of KRAS mutational status. MSI CSC lines carried mutations in genes coding for proteins with a role in RAS and calcium signaling, highlighting the role of a genomically unstable context in determining anti-EGFR resistance. Altogether, these results argue for a multifactorial origin of anti-EGFR resistance that emerges as the effect of multiple events targeting direct and indirect regulators of the EGFR pathway. An improved understanding of key molecular determinants of sensitivity/resistance to EGFR inhibition will be instrumental to optimize the clinical efficacy of anti-EGFR agents, representing a further step towards personalized treatments.
Collapse
Affiliation(s)
- Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Federica Francescangeli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Flavia Pucilli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Sara Vitale
- Institute of General Pathology, Catholic University and A. Gemelli Polyclinic, Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Marta Baiocchi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| |
Collapse
|
159
|
A mononucleotide repeat in PRRT2 is an important, frequent target of mismatch repair deficiency in cancer. Oncotarget 2018; 8:6043-6056. [PMID: 27907910 PMCID: PMC5351611 DOI: 10.18632/oncotarget.13464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 10/21/2016] [Indexed: 02/06/2023] Open
Abstract
The DNA mismatch repair (MMR) system corrects DNA replication mismatches thereby contributing to the maintenance of genomic stability. MMR deficiency has been observed in prostate cancer but its impact on the genomic landscape of these tumours is not known. In order to identify MMR associated mutations in prostate cancer we have performed whole genome sequencing of the MMR deficient PC346C prostate cancer cell line. We detected a total of 1196 mutations in PC346C which was 1.5-fold higher compared to a MMR proficient prostate cancer sample (G089). Of all different mutation classes, frameshifts in mononucleotide repeat (MNR) sequences were significantly enriched in the PC346C sample. As a result, a selection of genes with frameshift mutations in MNR was further assessed regarding its mutational status in a comprehensive panel of prostate, ovarian, endometrial and colorectal cancer cell lines. We identified PRRT2 and DAB2IP to be frequently mutated in MMR deficient cell lines, colorectal and endometrial cancer patient samples. Further characterization of PRRT2 revealed an important role of this gene in cancer biology. Both normal prostate cell lines and a colorectal cancer cell line showed increased proliferation, migration and invasion when expressing the mutated form of PRRT2 (ΔPRRT2). The wild-type PRRT2 (PRRT2wt) had an inhibitory effect in proliferation, consistent with the low expression level of PRRT2 in cancer versus normal prostate samples.
Collapse
|
160
|
Verma R, Sharma PC. Next generation sequencing-based emerging trends in molecular biology of gastric cancer. Am J Cancer Res 2018; 8:207-225. [PMID: 29511593 PMCID: PMC5835690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 11/29/2017] [Indexed: 06/08/2023] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer related mortality in the world. Being asymptomatic in nature till advanced stage, diagnosis of gastric cancer becomes difficult in early stages of the disease. The onset and progression of gastric cancer has been attributed to multiple factors including genetic alterations, epigenetic modifications, Helicobacter pylori and Epstein-Barr Virus (EBV) infection, and dietary habits. Next Generation Sequencing (NGS) based approaches viz. Whole Genome Sequencing (WGS), Whole Exome Sequencing (WES), RNA-Seq, and targeted sequencing have expanded the knowledge base of molecular pathogenesis of gastric cancer. In this review, we highlight recent NGS-based advances covering various genetic alterations (Microsatellite Instability, Single Nucleotide Variations, and Copy Number Variations), epigenetic changes (DNA methylation, histone modification, microRNAs) and differential gene expression during gastric tumorigenesis. We also briefly discuss the current and future potential biomarkers, drugs and therapeutic approaches available for the management of gastric cancer.
Collapse
Affiliation(s)
- Renu Verma
- University School of Biotechnology, Guru Gobind Singh Indraprastha UniversityNew Delhi 110078, India
| | - Prakash C Sharma
- University School of Biotechnology, Guru Gobind Singh Indraprastha UniversityNew Delhi 110078, India
| |
Collapse
|
161
|
Pan J, Lu L, Wang X, Liu D, Tian J, Liu H, Zhang M, Xu F, An F. AIM2 regulates vascular smooth muscle cell migration in atherosclerosis. Biochem Biophys Res Commun 2018; 497:401-409. [DOI: 10.1016/j.bbrc.2018.02.094] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/24/2022]
|
162
|
Fletcher R, Wang YJ, Schoen RE, Finn OJ, Yu J, Zhang L. Colorectal cancer prevention: Immune modulation taking the stage. Biochim Biophys Acta Rev Cancer 2018; 1869:138-148. [PMID: 29391185 DOI: 10.1016/j.bbcan.2017.12.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 02/07/2023]
Abstract
Prevention or early detection is one of the most promising strategies against colorectal cancer (CRC), the second leading cause of cancer death in the US. Recent studies indicate that antitumor immunity plays a key role in CRC prevention. Accumulating evidence suggests that immunosurveillance represents a critical barrier that emerging tumor cells have to overcome in order to sustain the course of tumor development. Virtually all of the agents with cancer preventive activity have been shown to have an immune modulating effect. A number of immunoprevention studies aimed at triggering antitumor immune response against early lesions have been performed, some of which have shown promising results. Furthermore, the recent success of immune checkpoint blockade therapy reinforces the notion that cancers including CRC can be effectively intervened via immune modulation including immune normalization, and has stimulated various immune-based combination prevention studies. This review summarizes recent advances to help better harness the immune system in CRC prevention.
Collapse
Affiliation(s)
- Rochelle Fletcher
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Yi-Jun Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Robert E Schoen
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Olivera J Finn
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lin Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA.
| |
Collapse
|
163
|
Lim CH, Cho YK, Kim SW, Choi MG, Rhee JK, Chung YJ, Lee SH, Kim TM. The chronological sequence of somatic mutations in early gastric carcinogenesis inferred from multiregion sequencing of gastric adenomas. Oncotarget 2018; 7:39758-39767. [PMID: 27175599 PMCID: PMC5129968 DOI: 10.18632/oncotarget.9250] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/26/2016] [Indexed: 12/26/2022] Open
Abstract
Mutation profiles and intratumoral heterogeneity are not well understood for benign gastric adenomas, some of which progress into malignant gastric adenocarcinomas. In this study, we performed whole-exome sequencing of three microsatellite stable (MSS) and two microsatellite instability-high (MSI-H) gastric adenomas with three regional tumor biopsies per case. We observed that the mutation abundance of benign gastric adenomas was comparable to those of gastric adenocarcinomas, suggesting that the mutational makeup for gastric carcinogenesis may already be achieved in benign adenomas. The extent of intratumoral heterogeneity was more substantial for MSS genomes in that only 1% - 14% of somatic mutations were common across the regional biopsies or 'public', while 50% - 94% of mutations were public in MSI-H gastric adenomas. We observed biallelic, loss-of-functional events of APC with truncating mutations and/or 5q losses for all cases, mostly as public events. All MSS gastric adenomas also harbored ARID2 truncating mutations, often as multiple, region-specific ones indicative of convergent evolution. Hotspot missense mutations on known cancer genes such as ERBB2 and KRAS were largely observed as region-specific aberrations. These findings suggest that biallelic functional APC inactivation initiates the gastric carcinogenesis and is followed by mutations of histone modifiers and then activation of known cancer-related genes. As the first exome-wide multi-region mutational profiling of gastric adenomas, our study provides clues on the chronological sequence of somatic mutations and their clonal architectures in early gastric carcinogenesis.
Collapse
Affiliation(s)
- Chul-Hyun Lim
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Yu Kyung Cho
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Sang Woo Kim
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Myung-Gyu Choi
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Je-Keun Rhee
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Yeun-Jun Chung
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Sug-Hyung Lee
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 137-701, South Korea
| |
Collapse
|
164
|
Stelloo E, Versluis MA, Nijman HW, de Bruyn M, Plat A, Osse EM, van Dijk RH, Nout RA, Creutzberg CL, de Bock GH, Smit VT, Bosse T, Hollema H. Microsatellite instability derived JAK1 frameshift mutations are associated with tumor immune evasion in endometrioid endometrial cancer. Oncotarget 2018; 7:39885-39893. [PMID: 27213585 PMCID: PMC5129978 DOI: 10.18632/oncotarget.9414] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/26/2016] [Indexed: 12/26/2022] Open
Abstract
JAK1 frameshift mutations may promote cancer cell immune evasion by impeding upregulation of the antigen presentation pathway in microsatellite unstable endometrial cancers (ECs). This study investigated the JAK1 mutation frequency, its functional implication in immune evasion and its prognostic significance in microsatellite unstable EC. Microsatellite instability and three microsatellite repeats within JAK1 were analyzed in 181 ECs. Sixty-two (34%) ECs showed microsatellite instability, of which 22 (35%) had a JAK1 mutation. LMP7, TAP1 and HLA class I protein expression and the presence of CD8-positive T-cells were analyzed in the microsatellite unstable ECs. JAK1 mutant microsatellite unstable ECs showed impaired upregulation of LMP7 (P=0.074) and HLA class I (P<0.001), validated using RNAseq data of the TCGA. TAP1 expression and presence of CD8-positive T-cells were not related to JAK1 mutations. In 198 additional microsatellite unstable ECs, the JAK1 mutation frequency was confirmed but no prognostic significance was found. For, JAK1 wildtype (n=135, 72%) and mutant (n=52, 28%) ECs, 10-year recurrence free rates were 84% and 77% (P=0.301). These observations show that JAK1 mutations are highly frequent in microsatellite unstable EC, not associated with survival, but are associated with impaired upregulation of LMP7 and HLA class I and may therefore facilitate immune escape.
Collapse
Affiliation(s)
- Ellen Stelloo
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco A Versluis
- Department of Gynecologic Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Hans W Nijman
- Department of Gynecologic Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Marco de Bruyn
- Department of Gynecologic Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Annechien Plat
- Department of Gynecologic Oncology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Elisabeth M Osse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Reinhardt H van Dijk
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Remi A Nout
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carien L Creutzberg
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Vincent T Smit
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Harry Hollema
- Department of Pathology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
165
|
Donahue TF, Bagrodia A, Audenet F, Donoghue MT, Cha EK, Sfakianos JP, Sperling D, Al-Ahmadie H, Clendenning M, Rosty C, Buchanan DD, Jenkins M, Hopper J, Winship I, Templeton AS, Walsh MF, Stadler ZK, Iyer G, Taylor B, Coleman J, Lindor NM, Solit DB, Bochner BH. Genomic Characterization of Upper-Tract Urothelial Carcinoma in Patients With Lynch Syndrome. JCO Precis Oncol 2018; 2018:PO.17.00143. [PMID: 30854504 PMCID: PMC6404976 DOI: 10.1200/po.17.00143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Patients with Lynch syndrome (LS) have a significantly increased risk of developing upper-tract urothelial carcinoma (UTUC). Here, we sought to identify differences in the patterns of mutational changes in LS-associated versus sporadic UTUCs. PATIENTS AND METHODS We performed targeted sequencing of 17 UTUCs from patients with documented LS-associated germline mutations (LS-UTUCs) using the Memorial Sloan Kettering Integrated Molecular Profiling of Actionable Cancer Targets targeted exon capture assay and compared the results with those from a recently characterized cohort of 82 patients with sporadic UTUC. RESULTS Patients with LS-UTUC were significantly younger, had had less exposure to tobacco, and more often presented with a ureteral primary site compared with patients with sporadic UTUC. The median number of mutations per tumor was significantly greater in LS-UTUC tumors than in tumors from the sporadic cohort (58; interquartile range [IQR], 47-101 v 6; IQR, 4-10; P < .001), as was the MSIsensor score (median, 25.1; IQR, 17.9-31.2 v 0.03; IQR, 0-0.44; P < .001). Differences in the genetic landscape were observed between sporadic and LS-associated tumors. Alterations in KMT2D, CREBBP, or ARID1A or in DNA damage response and repair genes were present at a significantly higher frequency in LS-UTUC. CIC, NOTCH1, NOTCH3, RB1, and CDKN1B alterations were almost exclusive to LS-UTUC. Although FGFR3 mutations were identified in both cohorts, the R248C hotspot mutation was highly enriched in LS-UTUC. CONCLUSION LSand sporadic UTUCs have overlapping but distinct genetic signatures. LS-UTUC is associated with hypermutation and a significantly higher prevalence of FGFR3 R248C mutation. Prospective molecular characterization of patients to identify those with LS-UTUC may help guide treatment.
Collapse
Affiliation(s)
- Timothy F. Donahue
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Aditya Bagrodia
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - François Audenet
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Mark T.A. Donoghue
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Eugene K. Cha
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - John P. Sfakianos
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Dahlia Sperling
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Hikmat Al-Ahmadie
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Mark Clendenning
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Christophe Rosty
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Daniel D. Buchanan
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Mark Jenkins
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - John Hopper
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Ingrid Winship
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Allyson S. Templeton
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Michael F. Walsh
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Zsofia K. Stadler
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Gopa Iyer
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Barry Taylor
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Jonathan Coleman
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Noralane M. Lindor
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - David B. Solit
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| | - Bernard H. Bochner
- Timothy F. Donahue, François Audenet, Mark T.A. Donoghue, Eugene K. Cha, Dahlia Sperling, Hikmat Al-Ahmadie, Michael F. Walsh, Zsofia K. Stadler, Gopa Iyer, Barry Taylor, Jonathan Coleman, David B. Solit, and Bernard H. Bochner, Memorial Sloan Kettering Cancer Center; John P. Sfakianos, Mount Sinai Hospital; Aditya Bagrodia, University of Texas Southwest Medical Center, Dallas, TX; Mark Clendenning, Christophe Rosty, Daniel D. Buchanan, Mark Jenkins, John Hopper, and Ingrid Winship, University of Melbourne, Parkville, Victoria, Australia; Allyson S. Templeton, Fred Hutchinson Cancer Research Center, Seattle, WA; and Noralane M. Lindor, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
166
|
Nagahashi M, Ling Y, Hayashida T, Kitagawa Y, Futamura M, Yoshida K, Kuwayama T, Nakamura S, Toshikawa C, Yamauchi H, Yamauchi T, Kaneko K, Kanbayashi C, Sato N, Miyoshi Y, Tsuchida J, Nakajima M, Shimada Y, Ichikawa H, Lyle S, Takabe K, Okuda S, Wakai T. Actionable gene alterations in an Asian population with triple-negative breast cancer. JCO Precis Oncol 2018; 2. [PMID: 32529167 DOI: 10.1200/po.17.00211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose It has been suggested that the biological characteristics of breast cancer may differ among different geographic or ethnic populations. Indeed, triple-negative breast cancer (TNBC), the most lethal breast cancer subgroup, has been reported to show a higher incidence in Japan than in the US. However, most genomic studies of these tumors are from Western countries and the genomic landscape of TNBC in an Asian population has not been thoroughly investigated. Here, we sought to elucidate the geographic and ethnic diversity of breast cancer by examining actionable driver alterations in TNBC tumors from Japanese patients and comparing them with The Cancer Genome Atlas (TCGA) database, which gather data primarily from non-Asian patients. Materials and Methods We performed comprehensive genomic profiling, including an analysis of 435 known cancer genes on Japanese TNBC patients (N=53) and compared the results to independent data obtained from TCGA (N=123). Results Driver alterations were identified in 51 out of 53 Japanese patients (96%). Although the overall alteration spectrum of Japanese patients was similar to that of the TCGA, we found significant differences in the frequencies of alterations in MYC and PTK2. We identified three patients (5.7%) with a high tumor mutation burden, although no microsatellite instability was observed in any of the Japanese patients. Importantly, pathway analysis revealed that 66.0% (35/53) of Japanese patients, as well as 66.7% (82/123) of the TCGA cohort, had alterations in at least one actionable gene targetable by an FDA-approved drug. Conclusion Our study identified actionable driver alterations in Japanese patients with TNBC, revealing new opportunities for targeted therapies in Asian patients.
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - YiWei Ling
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Manabu Futamura
- Department of Surgical Oncology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Takashi Kuwayama
- Division of Breast Surgical Oncology, Department of Surgery, Showa University School of Medicine, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Seigo Nakamura
- Division of Breast Surgical Oncology, Department of Surgery, Showa University School of Medicine, 1-5-8, Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Chie Toshikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan.,Department of Breast Surgical Oncology, Department of Internal Medicine, St. Luke's International Hospital, 9-1 Akashicho, Chuo, Tokyo 104-8560, Japan
| | - Hideko Yamauchi
- Department of Breast Surgical Oncology, Department of Internal Medicine, St. Luke's International Hospital, 9-1 Akashicho, Chuo, Tokyo 104-8560, Japan
| | - Teruo Yamauchi
- Division of Medical Oncology, Department of Internal Medicine, St. Luke's International Hospital, 9-1 Akashicho, Chuo, Tokyo 104-8560, Japan
| | - Koji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, 15-3 Kawagishi-cho 2-Chome, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Chizuko Kanbayashi
- Department of Breast Oncology, Niigata Cancer Center Hospital, 15-3 Kawagishi-cho 2-Chome, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, 15-3 Kawagishi-cho 2-Chome, Chuo-ku, Niigata City, Niigata 951-8566, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Junko Tsuchida
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Masato Nakajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Stephen Lyle
- University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, Massachusetts 01655, USA
| | - Kazuaki Takabe
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan.,Breast Surgery, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, New York 14263, USA.,Department of Surgery, University at Buffalo Jacobs School of Medicine and Biosciences, the State University of New York, USA.,Department of Breast Surgery and Oncology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan.,Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata 951-8510, Japan
| |
Collapse
|
167
|
Foltz SM, Liang WW, Xie M, Ding L. MIRMMR: binary classification of microsatellite instability using methylation and mutations. Bioinformatics 2017; 33:3799-3801. [PMID: 28961932 PMCID: PMC5860322 DOI: 10.1093/bioinformatics/btx507] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 08/03/2017] [Accepted: 08/09/2017] [Indexed: 01/27/2023] Open
Abstract
SUMMARY MIRMMR predicts microsatellite instability status in cancer samples using methylation and mutation information, in contrast to existing methods that rely on observed microsatellites. Additionally, MIRMMR highlights those genetic alterations contributing to microsatellite instability. AVAILABILITY AND IMPLEMENTATION Source code is freely available at https://github.com/ding-lab/MIRMMR under the MIT license, implemented in R and supported on Unix/OS X operating systems. CONTACT smfoltz@wustl.edu or lding@wustl.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Steven M Foltz
- Oncology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Wen-Wei Liang
- Oncology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Mingchao Xie
- Oncology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Li Ding
- Oncology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
168
|
Rhee JK, Jung YC, Kim KR, Yoo J, Kim J, Lee YJ, Ko YH, Lee HH, Cho BC, Kim TM. Impact of Tumor Purity on Immune Gene Expression and Clustering Analyses across Multiple Cancer Types. Cancer Immunol Res 2017; 6:87-97. [PMID: 29141981 DOI: 10.1158/2326-6066.cir-17-0201] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/30/2017] [Accepted: 11/08/2017] [Indexed: 11/16/2022]
Abstract
Surgical archives of tumor specimens are often impure. The presence of RNA transcripts from nontumor cells, such as immune and stromal cells, can impede analyses of cancer expression profiles. To systematically analyze the impact of tumor purity, the gene expression profiles and tumor purities were obtained for 7,794 tumor specimens across 21 tumor types (available in The Cancer Genome Atlas consortium). First, we observed that genes with roles in immunity and oxidative phosphorylation were significantly inversely correlated and correlated with the tumor purity, respectively. The expression of genes implicated in immunotherapy and specific immune cell genes, along with the abundance of immune cell infiltrates, was substantially inversely correlated with tumor purity. This relationship may explain the correlation between immune gene expression and mutation burden, highlighting the need to account for tumor purity in the evaluation of expression markers obtained from bulk tumor transcriptome data. Second, examination of cluster membership of gene pairs, with or without controlling for tumor purity, revealed that tumor purity may have a substantial impact on gene clustering across tumor types. Third, feature genes for molecular taxonomy were analyzed for correlation with tumor purity, and for some tumor types, feature genes representing the mesenchymal and classical subtypes were inversely correlated and correlated with tumor purity, respectively. Our findings indicate that tumor purity is an important confounder in evaluating the correlation between gene expression and clinicopathologic features such as mutation burden, as well as gene clustering and molecular taxonomy. Cancer Immunol Res; 6(1); 87-97. ©2017 AACR.
Collapse
Affiliation(s)
- Je-Keun Rhee
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yu Chae Jung
- Department of IT Engineering, Sookmyung Women's University, Seoul, Korea
| | - Kyu Ryung Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jinseon Yoo
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeeyoon Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Jae Lee
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon Ho Ko
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Han Hong Lee
- Division of Gastrointestinal Surgery, Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Korea.
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
169
|
Risk of colorectal cancer for carriers of a germ-line mutation in POLE or POLD1. Genet Med 2017; 20:890-895. [PMID: 29120461 PMCID: PMC5943186 DOI: 10.1038/gim.2017.185] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/17/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Germ-line mutations in the exonuclease domains of the POLE and POLD1 genes are associated with an increased, but yet unquantified, risk of colorectal cancer (CRC). METHODS We identified families with POLE or POLD1 variants by searching PubMed for relevant studies prior to October 2016 and by genotyping 669 population-based CRC cases diagnosed in patients under 60 years of age, from the Australasian Colorectal Cancer Family Registry. We estimated the age-specific cumulative risks (penetrance) using a modified segregation analysis. RESULTS We observed 67 CRCs (mean age at diagnosis = 50.2 (SD = 13.8) years) among 364 first- and second-degree relatives from 41 POLE families, and 6 CRCs (mean age at diagnosis = 39.7 (SD = 6.83) years) among 69 relatives from 9 POLD1 families. We estimated risks of CRC up to the age of 70 years (95% confidence interval) for males and females, respectively, to be 28% (95% CI, 10–42%) and 21% (95% CI, 7–33%) for POLE mutation carriers and 90% (95% CI, 33–99%) and 82% (95% CI, 26–99%) for POLD1 mutation carriers. CONCLUSION CRC risks for POLE mutation carriers are sufficiently high to warrant consideration of colonoscopy screening and implementation of management guidelines recommended for MSH6 mutation carriers in cases of Lynch syndrome. Refinement of estimates of CRC risk for POLD1 carriers is needed; however, clinical management recommendations could follow those made for POLE carriers.
Collapse
|
170
|
Albacker LA, Wu J, Smith P, Warmuth M, Stephens PJ, Zhu P, Yu L, Chmielecki J. Loss of function JAK1 mutations occur at high frequency in cancers with microsatellite instability and are suggestive of immune evasion. PLoS One 2017; 12:e0176181. [PMID: 29121062 PMCID: PMC5679612 DOI: 10.1371/journal.pone.0176181] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 04/06/2017] [Indexed: 12/17/2022] Open
Abstract
Immune evasion is a well-recognized hallmark of cancer and recent studies with immunotherapy agents have suggested that tumors with increased numbers of neoantigens elicit greater immune responses. We hypothesized that the immune system presents a common selective pressure on high mutation burden tumors and therefore immune evasion mutations would be enriched in high mutation burden tumors. The JAK family of kinases is required for the signaling of a host of immune modulators in tumor, stromal, and immune cells. Therefore, we analyzed alterations in this family for the hypothesized signature of an immune evasion mutation. Here, we searched a database of 61,704 unique solid tumors for alterations in the JAK family kinases (JAK1/2/3, TYK2). We used The Cancer Genome Atlas and Cancer Cell Line Encyclopedia data to confirm and extend our findings by analyzing gene expression patterns. Recurrent frameshift mutations in JAK1 were associated with high mutation burden and microsatellite instability. These mutations occurred in multiple tumor types including endometrial, colorectal, stomach, and prostate carcinomas. Analyzing gene expression signatures in endometrial and stomach adenocarcinomas revealed that tumors with a JAK1 frameshift exhibited reduced expression of interferon response signatures and multiple anti-tumor immune signatures. Importantly, endometrial cancer cell lines exhibited similar gene expression changes that were expected to be tumor cell intrinsic (e.g. interferon response) but not those expected to be tumor cell extrinsic (e.g. NK cells). From these data, we derive two primary conclusions: 1) JAK1 frameshifts are loss of function alterations that represent a potential pan-cancer adaptation to immune responses against tumors with microsatellite instability; 2) The mechanism by which JAK1 loss of function contributes to tumor immune evasion is likely associated with loss of the JAK1-mediated interferon response.
Collapse
Affiliation(s)
- Lee A. Albacker
- Foundation Medicine Inc., Cambridge, Massachusetts, United States of America
- * E-mail: (LA); (LY)
| | - Jeremy Wu
- H3 Biomedicine, Cambridge, Massachusetts, United States of America
| | - Peter Smith
- H3 Biomedicine, Cambridge, Massachusetts, United States of America
| | - Markus Warmuth
- H3 Biomedicine, Cambridge, Massachusetts, United States of America
| | - Philip J. Stephens
- Foundation Medicine Inc., Cambridge, Massachusetts, United States of America
| | - Ping Zhu
- H3 Biomedicine, Cambridge, Massachusetts, United States of America
| | - Lihua Yu
- H3 Biomedicine, Cambridge, Massachusetts, United States of America
- * E-mail: (LA); (LY)
| | - Juliann Chmielecki
- Foundation Medicine Inc., Cambridge, Massachusetts, United States of America
| |
Collapse
|
171
|
Frejno M, Zenezini Chiozzi R, Wilhelm M, Koch H, Zheng R, Klaeger S, Ruprecht B, Meng C, Kramer K, Jarzab A, Heinzlmeir S, Johnstone E, Domingo E, Kerr D, Jesinghaus M, Slotta-Huspenina J, Weichert W, Knapp S, Feller SM, Kuster B. Pharmacoproteomic characterisation of human colon and rectal cancer. Mol Syst Biol 2017; 13:951. [PMID: 29101300 PMCID: PMC5731344 DOI: 10.15252/msb.20177701] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Most molecular cancer therapies act on protein targets but data on the proteome status of patients and cellular models for proteome‐guided pre‐clinical drug sensitivity studies are only beginning to emerge. Here, we profiled the proteomes of 65 colorectal cancer (CRC) cell lines to a depth of > 10,000 proteins using mass spectrometry. Integration with proteomes of 90 CRC patients and matched transcriptomics data defined integrated CRC subtypes, highlighting cell lines representative of each tumour subtype. Modelling the responses of 52 CRC cell lines to 577 drugs as a function of proteome profiles enabled predicting drug sensitivity for cell lines and patients. Among many novel associations, MERTK was identified as a predictive marker for resistance towards MEK1/2 inhibitors and immunohistochemistry of 1,074 CRC tumours confirmed MERTK as a prognostic survival marker. We provide the proteomic and pharmacological data as a resource to the community to, for example, facilitate the design of innovative prospective clinical trials.
Collapse
Affiliation(s)
- Martin Frejno
- Department of Oncology, University of Oxford, Oxford, UK.,Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Riccardo Zenezini Chiozzi
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,Department of Chemistry, Sapienza - Università di Roma, Rome, Italy
| | - Mathias Wilhelm
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Heiner Koch
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Runsheng Zheng
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Susan Klaeger
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benjamin Ruprecht
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,Center for Integrated Protein Science (CIPSM), Munich, Germany
| | - Chen Meng
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Karl Kramer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Anna Jarzab
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Stephanie Heinzlmeir
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Enric Domingo
- Department of Oncology, University of Oxford, Oxford, UK.,Wellcome Trust Centre for Human Genetics (WTCHG), University of Oxford, Oxford, UK
| | - David Kerr
- Nuffield Division of Clinical Laboratory Sciences (NDCLS), University of Oxford, Oxford, UK
| | - Moritz Jesinghaus
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | | | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
| | - Stephan M Feller
- Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK .,Institute of Molecular Medicine, Martin-Luther-University, Halle, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany .,German Cancer Consortium (DKTK), Munich, Germany.,Center for Integrated Protein Science (CIPSM), Munich, Germany.,Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), Freising, Germany
| |
Collapse
|
172
|
Hertweck KL, Dasgupta S. The Landscape of mtDNA Modifications in Cancer: A Tale of Two Cities. Front Oncol 2017; 7:262. [PMID: 29164061 PMCID: PMC5673620 DOI: 10.3389/fonc.2017.00262] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/18/2017] [Indexed: 12/25/2022] Open
Abstract
Mitochondria from normal and cancerous cells represent a tale of two cities, wherein both execute similar processes but with different cellular and molecular effects. Given the number of reviews currently available which describe the functional implications of mitochondrial mutations in cancer, this article focuses on documenting current knowledge in the abundance and distribution of somatic mitochondrial mutations, followed by elucidation of processes which affect the fate of mutations in cancer cells. The conclusion includes an overview of translational implications for mtDNA mutations, as well as recommendations for future research uniting mitochondrial variants and tumorigenesis.
Collapse
Affiliation(s)
- Kate L Hertweck
- Department of Biology, The University of Texas at Tyler, Tyler, TX, United States
| | - Santanu Dasgupta
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| |
Collapse
|
173
|
Harada K, Kaya DM, Song S, Baba H, Ajani JA. Genomic profiling of colorectal cancers and the future of personalized treatment. COLORECTAL CANCER 2017. [DOI: 10.2217/crc-2016-0017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
New technologies have enabled faster, cheaper and more accurate genomic and other types of profiling. Therefore, treatment has become more customized according to molecular subtype. Here, we summarize the current status of genomic profiling for colorectal cancer (CRC) and discuss future directions. Recently, the CRC Subtyping Consortium classified CRC into four subtypes: CMS1, microsatellite instability immune (14%); CMS2, canonical (37%); CMS3, metabolic (13%); and CMS4, mesenchymal (23%). Testing for KRAS, NRAS and BRAF mutations, and microsatellite instability status in CRC has proven essential for treatment decisions. Tumor heterogeneity and the evolution of drug-resistant subclones after therapy should be further assessed and pursued. Patient-derived xenografts and liquid biopsies might facilitate the development of optimum and accurate personalized therapy regimens.
Collapse
Affiliation(s)
- Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1–1–1 Honjo, Kumamoto 860–8556, Japan
| | - Dilsa Mizrak Kaya
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, 1–1–1 Honjo, Kumamoto 860–8556, Japan
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
174
|
Shen T, Chen Z, Zhao ZJ, Wu J. Genetic defects of the IRF1-mediated major histocompatibility complex class I antigen presentation pathway occur prevalently in the JAK2 gene in non-small cell lung cancer. Oncotarget 2017; 8:60975-60986. [PMID: 28977839 PMCID: PMC5617399 DOI: 10.18632/oncotarget.17689] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/06/2017] [Indexed: 01/05/2023] Open
Abstract
Recognition of major histocompatibility complex (MHC) class I antigens on tumor cells by cytotoxic T cells is involved in T cell-mediated tumor immune surveillance and immune checkpoint therapy. The interferon-γ (IFNγ)-IRF1 signaling pathway regulates MHC class I antigen presentation. To examine genetic defects of the IFNγ-IRF1 pathway in non-small cell lung cancer (NSCLC), we analyzed The Cancer Genome Atlas (TCGA) lung adenocarcinoma (LuAd) and lung squamous cell carcinoma (LuSc) data. Loss-of-function (LOF) genetic alterations of the IFNγ-IRF1 pathway genes (IFNGR1, IFNGR2, JAK1, JAK2, STAT1, IRF1) were found in 64 (6.3%) of 1,016 patients. These genetic defects occur prevalently in JAK2 (33 cases) and often through deletions (29 cases) of chromosome 9p24.1. JAK2 deletions were frequently, but not always, associated with deletions of PD-L1 gene (CD274), PD-L2 gene (PDCD1LG2), PTPRD, and CDKN2A/CDKN2B at the chromosome 9p24.1-9p21.3 region. IRF1 expression was correlated with immune cytolytic activity markers GZMA and PRF1 in NSCLC. IFNγ induced IRF1 expression and cell surface HLA-A/HLA-B/HLA-C (HLA-ABC) in A549, H661, H292, and H2172 cells that contained the wildtype JAK2, but not in H1573 and H1623 cells that were JAK2 defective. Deletion of JAK2 or inhibition of the JAK2 kinase activity resulted in loss of IFNγ-induced IRF1 and cell surface HLA-ABC in JAK2 wildtype NSCLC cells, whereas expression of exogenous JAK2 in H1573 cells restored the IFNγ responses. These findings show that JAK2 deficiency is the major mechanism of genetic defects of the IFNγ-IRF1 pathway in NSCLC and reveal a previously unrecognized significance of chromosome 9p deletion in NSCLC.
Collapse
Affiliation(s)
- Tao Shen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Zhengming Chen
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, New York, USA
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Jie Wu
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
175
|
Analysis of somatic microsatellite indels identifies driver events in human tumors. Nat Biotechnol 2017; 35:951-959. [DOI: 10.1038/nbt.3966] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 08/18/2017] [Indexed: 01/03/2023]
|
176
|
Binder H, Hopp L, Schweiger MR, Hoffmann S, Jühling F, Kerick M, Timmermann B, Siebert S, Grimm C, Nersisyan L, Arakelyan A, Herberg M, Buske P, Loeffler-Wirth H, Rosolowski M, Engel C, Przybilla J, Peifer M, Friedrichs N, Moeslein G, Odenthal M, Hussong M, Peters S, Holzapfel S, Nattermann J, Hueneburg R, Schmiegel W, Royer-Pokora B, Aretz S, Kloth M, Kloor M, Buettner R, Galle J, Loeffler M. Genomic and transcriptomic heterogeneity of colorectal tumours arising in Lynch syndrome. J Pathol 2017; 243:242-254. [DOI: 10.1002/path.4948] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/01/2017] [Accepted: 07/14/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Hans Binder
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Lydia Hopp
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Michal R Schweiger
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Steve Hoffmann
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Frank Jühling
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques; Strasbourg France
- Université de Strasbourg; Strasbourg France
| | - Martin Kerick
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | | | - Susann Siebert
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Christina Grimm
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Lilit Nersisyan
- Group of Bioinformatics, Institute of Molecular Biology; National Academy of Sciences; Yerevan Armenia
| | - Arsen Arakelyan
- Group of Bioinformatics, Institute of Molecular Biology; National Academy of Sciences; Yerevan Armenia
| | - Maria Herberg
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Peter Buske
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Maciej Rosolowski
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University; Leipzig Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University; Leipzig Germany
| | - Jens Przybilla
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Martin Peifer
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Nicolaus Friedrichs
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Gabriela Moeslein
- Department of Hereditary Tumour Syndromes; Surgical Centre, HELIOS Clinic, University Witten/Herdecke; Wuppertal Germany
| | - Margarete Odenthal
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Michelle Hussong
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
- Translational Epigenomics; University Hospital Cologne; Cologne Germany
- Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Sophia Peters
- Institute of Human Genetics, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Stefanie Holzapfel
- Institute of Human Genetics, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Robert Hueneburg
- Department of Internal Medicine I, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Wolff Schmiegel
- Department of Medicine, Haematology and Oncology; Ruhr-University of Bochum, Knappschaftskrankenhaus; Bochum Germany
| | - Brigitte Royer-Pokora
- Institute of Human Genetics and Anthropology; Heinrich-Heine University; Düsseldorf Germany
| | - Stefan Aretz
- Institute of Human Genetics, University Hospital Bonn; Centre for Hereditary Tumour Syndromes, University of Bonn; Bonn Germany
| | - Michael Kloth
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Matthias Kloor
- Department of Applied Tumour Biology, Institute of Pathology; University Hospital Heidelberg; Heidelberg Germany
- Clinical Cooperation Unit of Applied Tumour Biology; DKFZ (German Cancer Research Centre) Heidelberg; Germany
- Molecular Medicine Partnership Unit; University Hospital Heidelberg and EMBL Heidelberg; Heidelberg Germany
| | - Reinhard Buettner
- Institute of Pathology, Centre for Integrated Oncology; University Hospital Cologne; Cologne Germany
| | - Jörg Galle
- Interdisciplinary Centre for Bioinformatics; Leipzig University; Leipzig Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University; Leipzig Germany
| |
Collapse
|
177
|
Hypermutated tumours in the era of immunotherapy: The paradigm of personalised medicine. Eur J Cancer 2017; 84:290-303. [PMID: 28846956 DOI: 10.1016/j.ejca.2017.07.026] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors have demonstrated unprecedented clinical activity in a wide range of cancers. Significant therapeutic responses have recently been observed in patients presenting mismatch repair-deficient (MMRD) tumours. MMRD cancers exhibit a remarkably high rate of mutations, which can result in the formation of neoantigens, hypothesised to enhance the antitumour immune response. In addition to MMRD tumours, cancers mutated in the exonuclease domain of the catalytic subunit of the DNA polymerase epsilon (POLE) also exhibit an ultramutated genome and are thus likely to benefit from immunotherapy. In this review, we provide an overview of recent data on hypermutated tumours, including MMRD and POLE-mutated cancers, with a focus on their distinctive clinicopathological and molecular characteristics as well as their immune environment. We also discuss the emergence of immune therapy to treat these hypermutated cancers, and we comment on the recent Food and Drug Administration approval of an immune checkpoint inhibitor, the programmed cell death 1 antibody (pembrolizumab, Keytruda), for the treatment of patients with metastatic MMRD cancers regardless of the tumour type. This breakthrough represents a turning point in the management of these hypermutated tumours and paves the way for broader strategies in immunoprecision medicine.
Collapse
|
178
|
Qing Y, Gerson SL. Mismatch repair deficient hematopoietic stem cells are preleukemic stem cells. PLoS One 2017; 12:e0182175. [PMID: 28767666 PMCID: PMC5540588 DOI: 10.1371/journal.pone.0182175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/13/2017] [Indexed: 12/27/2022] Open
Abstract
Whereas transformation events in hematopoietic malignancies may occur at different developmental stages, the initial mutation originates in hematopoietic stem cells (HSCs), creating a preleukemic stem cell (PLSC). Subsequent mutations at either stem cell or progenitor cell levels transform the PLSC into lymphoma/leukemia initiating cells (LIC). Thymic lymphomas have been thought to develop from developing thymocytes. T cell progenitors are generated from HSCs in the bone marrow (BM), but maturation and proliferation of T cells as well as T-lymphomagenesis depends on both regulatory mechanisms and microenvironment within the thymus. We studied PLSC linked to thymic lymphomas. In this study, we use MSH2-/- mice as a model to investigate the existence of PLSC and the evolution of PLSC to LIC. Following BM transplantation, we found that MSH2-/- BM cells from young mice are able to fully reconstitute multiple hematopoietic lineages of lethally irradiated wild-type recipients. However, all recipients developed thymic lymphomas within three and four months post transplantation. Transplantation of different fractions of BM cells or thymocytes from young health MSH2-/- mice showed that an HSC enriched fraction always reconstituted hematopoiesis followed by lymphoma development. In addition, lymphomas did not occur in thymectomized recipients of MSH2-/- BM. These results suggest that HSCs with DNA repair defects such as MSH2-/- are PLSCs because they retain hematopoietic function, but also carry an obligate lymphomagenic potential within their T-cell progeny that is dependent on the thymic microenvironment.
Collapse
Affiliation(s)
- Yulan Qing
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Stanton L. Gerson
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
179
|
Endometrial cancer gene panels: clinical diagnostic vs research germline DNA testing. Mod Pathol 2017; 30:1048-1068. [PMID: 28452373 DOI: 10.1038/modpathol.2017.20] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/07/2017] [Accepted: 02/09/2017] [Indexed: 12/12/2022]
Abstract
Endometrial cancer is the most common gynecological cancer, but is nevertheless uncommon enough to have value as a signature cancer for some hereditary cancer syndromes. Commercial multigene testing panels include up to 13 different genes annotated for germline DNA testing of patients with endometrial cancer. Many other genes have been reported as relevant to familial endometrial cancer from directed genome-wide sequencing studies or multigene panel testing, or research. This review assesses the evidence supporting association with endometrial cancer risk for 32 genes implicated in hereditary endometrial cancer, and presents a summary of rare germline variants in these 32 genes detected by analysis of quasi-population-based endometrial cancer patients from The Cancer Genome Atlas project. This comprehensive investigation has led to the conclusion that convincing evidence currently exists to support clinical testing of only six of these genes for diagnosis of hereditary endometrial cancer. Testing of endometrial cancer patients for the remaining genes should be considered in the context of research studies, as a means to better establish the level of endometrial cancer risk, if any, associated with genetic variants that are deleterious to gene or protein function. It is acknowledged that clinical testing of endometrial cancer patients for several genes included on commercial panels may provide actionable findings in relation to risk of other cancers, but these should be considered secondary or incidental findings and not conclusive evidence for diagnosis of inherited endometrial cancer. In summary, this review and analysis provides a comprehensive report of current evidence to guide the selection of genes for clinical and research gene testing of germline DNA from endometrial cancer patients.
Collapse
|
180
|
Abstract
PURPOSE OF REVIEW Inflammasomes are major actors of the innate immune system, through their regulation of inflammatory caspases and maturation of IL-1β and IL-18. These multiprotein complexes have been shown to play major roles in inflammatory and metabolic diseases and have more recently been implicated in tumor development and dissemination. In this review, we address these recent findings, focusing particularly on colorectal cancer (CRC) initiation and tumor dissemination. RECENT FINDINGS Based mostly on loss-of-function experiments in mouse models, paradoxical results were obtained as both protumoral and antitumoral activities were reported. Moreover, several studies report major inflammasome-independent functions for some of these innate receptor proteins such as absent in melanoma 2, nod-like receptor family pyrin containing 3 (NLRP3) or nod-like receptor family CARD containing 4 (NLRC4), functions exerted in epithelial cells as well as in immune cells. SUMMARY The current review summarizes recent findings on the implication of inflammasomes and of absent in melanoma 2, NLRC4 and NLRP3 inflammasome-independent functions in cancer development and dissemination. Although contradictory in certain aspects, these studies highlight a lack of understanding of their mechanistic functions and regulations in cancer and the need for further investigations.
Collapse
|
181
|
Wang JS, Yan YH, Zhang DY. Modular probes for enriching and detecting complex nucleic acid sequences. Nat Chem 2017; 9:1222-1228. [PMID: 29168489 DOI: 10.1038/nchem.2820] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 06/06/2017] [Indexed: 01/28/2023]
Abstract
Complex DNA sequences are difficult to detect and profile, but are important contributors to human health and disease. Existing hybridization probes lack the capability to selectively bind and enrich hypervariable, long or repetitive sequences. Here, we present a generalized strategy for constructing modular hybridization probes (M-Probes) that overcomes these challenges. We demonstrate that M-Probes can tolerate sequence variations of up to 7 nt at prescribed positions while maintaining single nucleotide sensitivity at other positions. M-Probes are also shown to be capable of sequence-selectively binding a continuous DNA sequence of more than 500 nt. Furthermore, we show that M-Probes can detect genes with triplet repeats exceeding a programmed threshold. As a demonstration of this technology, we have developed a hybrid capture method to determine the exact triplet repeat expansion number in the Huntington's gene of genomic DNA using quantitative PCR.
Collapse
Affiliation(s)
- Juexiao Sherry Wang
- Systems, Synthetic, and Physical Biology, Rice University, Houston, Texas 77030, USA.,Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - Yan Helen Yan
- Systems, Synthetic, and Physical Biology, Rice University, Houston, Texas 77030, USA.,Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| | - David Yu Zhang
- Systems, Synthetic, and Physical Biology, Rice University, Houston, Texas 77030, USA.,Department of Bioengineering, Rice University, Houston, Texas 77030, USA
| |
Collapse
|
182
|
He L, Chen Y, Wu Y, Xu Y, Zhang Z, Liu Z. Nucleic acid sensing pattern recognition receptors in the development of colorectal cancer and colitis. Cell Mol Life Sci 2017; 74:2395-2411. [PMID: 28224203 PMCID: PMC11107753 DOI: 10.1007/s00018-017-2477-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 01/02/2017] [Accepted: 01/26/2017] [Indexed: 12/16/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths that is often associated with inflammation initiated by activation of pattern recognition receptors (PRRs). Nucleic acid sensing PRRs are one of the major subsets of PRRs that sense nucleic acid (DNA and RNA), mainly including some members of Toll-like receptors (TLR3, 7, 8, 9), AIM2-like receptors (AIM2, IFI16), STING, cGAS, RNA polymerase III, and DExD/H box nucleic acid helicases (such as RIG-I like receptors (RIG-I, MDA5, LPG2), DDX1, 3, 5, 7, 17, 21, 41, 60, and DHX9, 36). Activation of these receptors eventually leads to the release of cytokines and activation of immune cells, which are well known to play crucial roles in host defense against intracellular bacterial and virus infection. However, the functions of these nucleic acid sensing PRRs in the other diseases such as CRC and colitis remain largely unknown. Recent studies indicated that nucleic acid sensing PRRs contribute to CRC and/or colitis development, and therapeutic modulation of nucleic acid sensing PRRs may reduce the risk of CRC development. However, until now, a comprehensive review on the role of nucleic acid sensing PRRs in CRC and colitis is still lacking. This review provided an overview of the roles as well as the mechanisms of these nucleic acid sensing PRRs (AIM2, STING, cGAS, RIG-I and its downstream molecules, DDX3, 5, 6,17, and DHX9, 36) in CRC and colitis, which may aid the diagnosis, therapy, and prognostic prediction of CRC and colitis.
Collapse
Affiliation(s)
- Liangmei He
- Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yayun Chen
- Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Yuanbing Wu
- Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Ying Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Zixiang Zhang
- The First Affiliated Hospital of Gannan Medical University, Gannan Medical University, Ganzhou, 341000, Jiangxi, China.
| | - Zhiping Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
183
|
Kondelin J, Gylfe AE, Lundgren S, Tanskanen T, Hamberg J, Aavikko M, Palin K, Ristolainen H, Katainen R, Kaasinen E, Taipale M, Taipale J, Renkonen-Sinisalo L, Järvinen H, Böhm J, Mecklin JP, Vahteristo P, Tuupanen S, Aaltonen LA, Pitkänen E. Comprehensive Evaluation of Protein Coding Mononucleotide Microsatellites in Microsatellite-Unstable Colorectal Cancer. Cancer Res 2017; 77:4078-4088. [PMID: 28611049 DOI: 10.1158/0008-5472.can-17-0682] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/24/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Approximately 15% of colorectal cancers exhibit microsatellite instability (MSI), which leads to accumulation of large numbers of small insertions and deletions (indels). Genes that provide growth advantage to cells via loss-of-function mutations in microsatellites are called MSI target genes. Several criteria to define these genes have been suggested, one of them being simple mutation frequency. Microsatellite mutation rate, however, depends on the length and nucleotide context of the microsatellite. Therefore, assessing the general impact of mismatch repair deficiency on the likelihood of mutation events is paramount when following this approach. To identify MSI target genes, we developed a statistical model for the somatic background indel mutation rate of microsatellites to assess mutation significance. Exome sequencing data of 24 MSI colorectal cancers revealed indels at 54 million mononucleotide microsatellites of three or more nucleotides in length. The top 105 microsatellites from 71 genes were further analyzed in 93 additional MSI colorectal cancers. Mutation significance and estimated clonality of mutations determined the most likely MSI target genes to be the aminoadipate-semialdehyde dehydrogenase AASDH and the solute transporter SLC9A8 Our findings offer a systematic profiling of the somatic background mutation rate in protein-coding mononucleotide microsatellites, allowing a full cataloging of the true targets of MSI in colorectal cancer. Cancer Res; 77(15); 4078-88. ©2017 AACR.
Collapse
Affiliation(s)
- Johanna Kondelin
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Alexandra E Gylfe
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Sofie Lundgren
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Tomas Tanskanen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Jiri Hamberg
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Mervi Aavikko
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Kimmo Palin
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Heikki Ristolainen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Riku Katainen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Eevi Kaasinen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Minna Taipale
- Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden.,Science for Life Center, Huddinge, Sweden
| | - Jussi Taipale
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden.,Science for Life Center, Huddinge, Sweden
| | - Laura Renkonen-Sinisalo
- Department of Surgery, Helsinki University Central Hospital, Hospital District of Helsinki and Uusimaa, Helsinki, Finland
| | - Heikki Järvinen
- Department of Surgery, Helsinki University Central Hospital, Hospital District of Helsinki and Uusimaa, Helsinki, Finland
| | - Jan Böhm
- Department of Pathology, Jyväskylä Central Hospital, Jyväskylä, Finland
| | - Jukka-Pekka Mecklin
- Department of Surgery, Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä, Finland
| | - Pia Vahteristo
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Sari Tuupanen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Lauri A Aaltonen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland.,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Solna, Sweden
| | - Esa Pitkänen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland. .,Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| |
Collapse
|
184
|
A molecular portrait of microsatellite instability across multiple cancers. Nat Commun 2017; 8:15180. [PMID: 28585546 PMCID: PMC5467167 DOI: 10.1038/ncomms15180] [Citation(s) in RCA: 415] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/03/2017] [Indexed: 12/30/2022] Open
Abstract
Microsatellite instability (MSI) refers to the hypermutability of short repetitive sequences in the genome caused by impaired DNA mismatch repair. Although MSI has been studied for decades, large amounts of sequencing data now available allows us to examine the molecular fingerprints of MSI in greater detail. Here, we analyse ∼8,000 exomes and ∼1,000 whole genomes of cancer patients across 23 cancer types. Our analysis reveals that the frequency of MSI events is highly variable within and across tumour types. We also identify genes in DNA repair and oncogenic pathways recurrently subject to MSI and uncover non-coding loci that frequently display MSI. Finally, we propose a highly accurate exome-based predictive model for the MSI phenotype. These results advance our understanding of the genomic drivers and consequences of MSI, and our comprehensive catalogue of tumour-type-specific MSI loci will enable panel-based MSI testing to identify patients who are likely to benefit from immunotherapy. Some cancers with DNA mismatch repair deficiency display microsatellite instability. Here the authors analyse twenty three cancer types at the exome and whole-genome level, and identify loci with recurrent microsatellite instability that could be used to identify patients who would benefit from immunotherapy.
Collapse
|
185
|
Liu W, Qian K, Wei X, Deng H, Zhao B, Chen Q, Zhang J, Liu H. miR‑27a promotes proliferation, migration, and invasion of colorectal cancer by targeting FAM172A and acts as a diagnostic and prognostic biomarker. Oncol Rep 2017; 37:3554-3564. [PMID: 28440497 DOI: 10.3892/or.2017.5592] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/10/2017] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence shows that mircroRNAs (miRNAs) play a crucial role in the development of colorectal cancer. In our previous study, FAM172A was demonstrated to be a novel tumor suppressor gene in CRC. Therefore, the aim of the present study was to identify whether the miR‑27a could be a diagnostic and prognostic marker and the regulatory relationships between miR‑27a and FAM172A. We demonstrated high levels of miR‑27a expression in tissues of patients with CRC as well as in CRC cell lines. There was a positive correlation between the levels of miR‑27a and the poor overall survival of patients with CRC. Furthermore, elevated levels of miR‑27a expression were associated with TNM stage and distant metastasis. Increased expression or inhibition of miR‑27a promoted or inhibited the metastasis of CRC cell lines, respectively. Moreover, we showed that miR‑27a directly targets the 3'-untranslated region of FAM172A mRNA by using a dual-luciferase assay. Increased or decreased expression of FAM172A expression was observed when miR‑27a expression was inhibited or elevated in the CRC cells, respectively. In summary, our study showed that miR‑27a expression is a diagnostic and prognostic marker and correlates with overall survival of patients with CRC. Therefore, it may be a therapeutic approach for preventing metastasis of CRC to inhibit expression of miR‑27a or increase expression of FAM172A.
Collapse
Affiliation(s)
- Wenjun Liu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Kai Qian
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xing Wei
- The First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haijun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Bei Zhao
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qing Chen
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jinqian Zhang
- Department of Laboratory Medicine, The Second People's Hospital of Guangdong Province, Guangzhou, Guangdong 510317, P.R. China
| | - Hao Liu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
186
|
Sveen A, Johannessen B, Tengs T, Danielsen SA, Eilertsen IA, Lind GE, Berg KCG, Leithe E, Meza-Zepeda LA, Domingo E, Myklebost O, Kerr D, Tomlinson I, Nesbakken A, Skotheim RI, Lothe RA. Multilevel genomics of colorectal cancers with microsatellite instability-clinical impact of JAK1 mutations and consensus molecular subtype 1. Genome Med 2017; 9:46. [PMID: 28539123 PMCID: PMC5442873 DOI: 10.1186/s13073-017-0434-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/03/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Approximately 15% of primary colorectal cancers have DNA mismatch repair deficiency, causing a complex genome with thousands of small mutations-the microsatellite instability (MSI) phenotype. We investigated molecular heterogeneity and tumor immunogenicity in relation to clinical endpoints within this distinct subtype of colorectal cancers. METHODS A total of 333 primary MSI+ colorectal tumors from multiple cohorts were analyzed by multilevel genomics and computational modeling-including mutation profiling, clonality modeling, and neoantigen prediction in a subset of the tumors, as well as gene expression profiling for consensus molecular subtypes (CMS) and immune cell infiltration. RESULTS Novel, frequent frameshift mutations in four cancer-critical genes were identified by deep exome sequencing, including in CRTC1, BCL9, JAK1, and PTCH1. JAK1 loss-of-function mutations were validated with an overall frequency of 20% in Norwegian and British patients, and mutated tumors had up-regulation of transcriptional signatures associated with resistance to anti-PD-1 treatment. Clonality analyses revealed a high level of intra-tumor heterogeneity; however, this was not associated with disease progression. Among the MSI+ tumors, the total mutation load correlated with the number of predicted neoantigens (P = 4 × 10-5), but not with immune cell infiltration-this was dependent on the CMS class; MSI+ tumors in CMS1 were highly immunogenic compared to MSI+ tumors in CMS2-4. Both JAK1 mutations and CMS1 were favorable prognostic factors (hazard ratios 0.2 [0.05-0.9] and 0.4 [0.2-0.9], respectively, P = 0.03 and 0.02). CONCLUSIONS Multilevel genomic analyses of MSI+ colorectal cancer revealed molecular heterogeneity with clinical relevance, including tumor immunogenicity and a favorable patient outcome associated with JAK1 mutations and the transcriptomic subgroup CMS1, emphasizing the potential for prognostic stratification of this clinically important subtype. See related research highlight by Samstein and Chan 10.1186/s13073-017-0438-9.
Collapse
Affiliation(s)
- Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Torstein Tengs
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Stine A. Danielsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Ina A. Eilertsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Guro E. Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Kaja C. G. Berg
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Leonardo A. Meza-Zepeda
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Genomics Core Facility, Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
| | - Enric Domingo
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Ola Myklebost
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
| | - David Kerr
- Department of Oncology, University of Oxford, Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Ian Tomlinson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN UK
| | - Arild Nesbakken
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
- Department of Gastrointestinal Surgery, Oslo University Hospital, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Rolf I. Skotheim
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| | - Ragnhild A. Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Norwegian Cancer Genomics Consortium, Oslo University Hospital, P.O. Box 4953, Nydalen, NO-0424 Oslo Norway
- Centre for Cancer Biomedicine, Institute for Clinical Medicine, University of Oslo, P.O. Box 4950, Nydalen, NO-0424 Oslo Norway
| |
Collapse
|
187
|
The current value of determining the mismatch repair status of colorectal cancer: A rationale for routine testing. Crit Rev Oncol Hematol 2017; 116:38-57. [PMID: 28693799 DOI: 10.1016/j.critrevonc.2017.05.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/02/2017] [Accepted: 05/15/2017] [Indexed: 02/08/2023] Open
Abstract
Colorectal Cancer (CRC) is the third most prevalent cancer in men and women. Up to 15% of CRCs display microsatellite instability (MSI). MSI is reflective of a deficient mismatch repair (MMR) system and is most commonly caused by hypermethylation of the MLH1 promoter. However, it may also be due to autosomal dominant constitutional mutations in DNA MMR, termed Lynch Syndrome. MSI may be diagnosed via polymerase chain reaction (PCR) or alternatively, immunohistochemistry (IHC) can identify MMR deficiency (dMMR). Many institutions now advocate universal tumor screening of CRC via either PCR for MSI or IHC for dMMR to guide Lynch Syndrome testing. The association of sporadic MSI with methylation of the MLH1 promoter and an activating BRAF mutation may offer further exclusion criteria for genetic testing. Aside from screening for Lynch syndrome, MMR testing is important because of its prognostic and therapeutic implications. Several studies have shown MSI CRCs exhibit different clinicopathological features and prognosis compared to microsatellite-stable (MSS) CRCs. For example, response to conventional chemotherapy has been reported to be less in MSI tumours. More recently, MSI tumours have been shown to be responsive to immune-checkpoint inhibition providing a novel therapeutic strategy. This provides a rationale for routine testing for MSI or dMMR in CRC.
Collapse
|
188
|
Libera L, Sahnane N, Carnevali IW, Cimetti L, Cerutti R, Chiaravalli AM, Riva C, Tibiletti MG, Sessa F, Furlan D. Microsatellite analysis of sporadic and hereditary gynaecological cancer in routine diagnostics. J Clin Pathol 2017; 70:792-797. [DOI: 10.1136/jclinpath-2017-204348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 11/04/2022]
Abstract
Microsatellite instability (MSI) testing is tricky in gynaecological cancers (GC). Thus, we aimed to describe the instability patterns to improve MSI test interpretation in sporadic and hereditary GCs. Ninety-five cases, including uterine and ovarian cancers, with known genetic and immunohistochemical (IHC) features, were analysed for MSI by a mononucleotide repeats pentaplex (MRP). We identified 13 ambiguous cases that did not fully meet MSI criteria (‘borderline’ cases, B-MSI), which were mainly represented by MSH2/MSH6-deficient and Lynch syndrome cases. Also, we evaluated nine additional loci of candidate MSI markers that did not improve the detection of MSI cases, but might be useful for discordant or borderline samples. In conclusion, although MSI and IHC test are highly concordant, a subset of ambiguous MSI cases deserves a careful interpretation in particular when MSH2/MSH6 are involved.RPL22andSRPRtesting may be useful to integrate MRP panel for the analysis of critical cases.
Collapse
|
189
|
Fricke F, Lee J, Michalak M, Warnken U, Hausser I, Suarez-Carmona M, Halama N, Schnölzer M, Kopitz J, Gebert J. TGFBR2-dependent alterations of exosomal cargo and functions in DNA mismatch repair-deficient HCT116 colorectal cancer cells. Cell Commun Signal 2017; 15:14. [PMID: 28376875 PMCID: PMC5379773 DOI: 10.1186/s12964-017-0169-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/21/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Colorectal cancers (CRCs) that lack DNA mismatch repair function exhibit the microsatellite unstable (MSI) phenotype and are characterized by the accumulation of frameshift mutations at short repetitive DNA sequences (microsatellites). These tumors recurrently show inactivating frameshift mutations in the tumor suppressor Transforming Growth Factor Beta Receptor Type 2 (TGFBR2) thereby abrogating downstream signaling. How altered TGFBR2 signaling affects exosome-mediated communication between MSI tumor cells and their environment has not been resolved. Here, we report on molecular alterations of exosomes shed by MSI cells and the biological response evoked in recipient cells. METHODS Exosomes were isolated and characterized by electron microscopy, nanoparticle tracking, and western blot analysis. TGFBR2-dependent effects on the cargo and functions of exosomes were studied in a MSI CRC model cell line enabling reconstituted and inducible TGFBR2 expression and signaling. Microsatellite frameshift mutations in exosomal and cellular DNA were examined by PCR-based DNA fragment analysis and exosomal protein profiles were identified by mass spectrometry. Uptake of fluorescent-labeled exosomes by hepatoma recipient cells was monitored by confocal microscopy. TGFBR2-dependent exosomal effects on secreted cytokine levels of recipient cells were analyzed by Luminex technology and ELISA. RESULTS Frameshift mutation patterns in microsatellite stretches of TGFBR2 and other MSI target genes were found to be reflected in the cargo of MSI CRC-derived exosomes. At the proteome level, reconstituted TGFBR2 expression and signaling uncovered two protein subsets exclusively occurring in exosomes derived from TGFBR2-deficient (14 proteins) or TGFBR2-proficient (five proteins) MSI donor cells. Uptake of these exosomes by recipient cells caused increased secretion (2-6 fold) of specific cytokines (Interleukin-4, Stem Cell Factor, Platelet-derived Growth Factor-B), depending on the TGFBR2 expression status of the tumor cell. CONCLUSION Our results indicate that the coding MSI phenotype of DNA mismatch repair-deficient CRC cells is maintained in their exosomal DNA. Moreover, we uncovered that a recurrent MSI tumor driver mutation like TGFBR2 can reprogram the protein content of MSI cell-derived exosomes and in turn modulate the cytokine secretion profile of recipient cells. Apart from its diagnostic potential, these TGFBR2-dependent exosomal molecular and proteomic signatures might help to understand the signaling routes used by MSI tumors. Fricke et al. uncovered coding microsatellite instability-associated mutations of colorectal tumor driver genes like TGFBR2 in MSI tumor cellderived exosomes. Depending on the TGFBR2 expression status of their donor cells, shed exosomes show distinct proteomic signatures and promote altered cytokine secretion profiles in recipient cells.
Collapse
Affiliation(s)
- Fabia Fricke
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Department of Cancer Early Detection, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Jennifer Lee
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Department of Cancer Early Detection, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Present address: Tissue Genesis, Suite 1000, Tissue Genesis Tower, 810 Richards Street, Honolulu, HI 96813 USA
| | - Malwina Michalak
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Department of Cancer Early Detection, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Uwe Warnken
- Functional Proteome Analysis and Core Facility Protein Analysis (B100), German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Ingrid Hausser
- Department of General Pathology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Meggy Suarez-Carmona
- Department of Medical Oncology, National Center for Tumor diseases (NCT), Tissue Imaging and Analysis Center, Bioquant, University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor diseases (NCT), Tissue Imaging and Analysis Center, Bioquant, University Hospital Heidelberg, Im Neuenheimer Feld 460, 69120 Heidelberg, Germany
| | - Martina Schnölzer
- Functional Proteome Analysis and Core Facility Protein Analysis (B100), German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jürgen Kopitz
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Department of Cancer Early Detection, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Johannes Gebert
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
- Department of Cancer Early Detection, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| |
Collapse
|
190
|
|
191
|
Buccitelli C, Salgueiro L, Rowald K, Sotillo R, Mardin BR, Korbel JO. Pan-cancer analysis distinguishes transcriptional changes of aneuploidy from proliferation. Genome Res 2017; 27:501-511. [PMID: 28320919 PMCID: PMC5378169 DOI: 10.1101/gr.212225.116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/27/2017] [Indexed: 01/09/2023]
Abstract
Patterns of gene expression in tumors can arise as a consequence of or result in genomic instability, characterized by the accumulation of somatic copy number alterations (SCNAs) and point mutations (PMs). Expression signatures have been widely used as markers for genomic instability, and both SCNAs and PMs could be thought to associate with distinct signatures given their different formation mechanisms. Here we test this notion by systematically investigating SCNA, PM, and transcriptome data from 2660 cancer patients representing 11 tumor types. Notably, our data indicate that similar expression signatures can be derived from correlating gene expression with either SCNA or PM load. Gene sets related to cell growth and proliferation generally associated positively, and immunoregulatory gene sets negatively, with variant burden. In-depth analyses revealed several genes whose de-regulation correlates with SCNA but not with PM burden, yielding downstream effectors of TP53 and MYC signaling unique to high-SCNA tumors. We compared our findings to expression changes observed in two different cancer mouse models with persistent mitotic chromosomal instability, observing a decrease in proliferative expression signatures. Our results suggest that overexpression of cell-cycle-related genes are a characteristic of proliferation, and likely tumor evolution, rather than ongoing genomic instability.
Collapse
Affiliation(s)
- Christopher Buccitelli
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany
| | - Lorena Salgueiro
- Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Konstantina Rowald
- Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Rocio Sotillo
- Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Balca R Mardin
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany
| | - Jan O Korbel
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany.,EMBL-European Bioinformatics Institute (EMBL-EBI), Hinxton CB10 1SD, United Kingdom
| |
Collapse
|
192
|
Loeb LA. Human Cancers Express a Mutator Phenotype: Hypothesis, Origin, and Consequences. Cancer Res 2017; 76:2057-9. [PMID: 27197248 DOI: 10.1158/0008-5472.can-16-0794] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 11/16/2022]
Abstract
The mutator phenotype hypothesis was postulated more than 40 years ago. It was based on the multiple enzymatic steps required to precisely replicate the 6 billion bases in the human genome each time a normal cell divides. A reduction in this accuracy during tumor progression could be responsible for the striking heterogeneity of malignant cells within a tumor and for the rapidity by which cancers become resistant to therapy. Cancer Res; 76(8); 2057-9. ©2016 AACRSee related article by Loeb et al. Cancer Res. 1974;34:2311-21.
Collapse
Affiliation(s)
- Lawrence A Loeb
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington. Department of Biochemistry, University of Washington School of Medicine, Seattle, Washington.
| |
Collapse
|
193
|
Immune checkpoint inhibition and its relationship with hypermutation phenoytype as a potential treatment for Glioblastoma. J Neurooncol 2017; 132:359-372. [PMID: 28293764 DOI: 10.1007/s11060-017-2390-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 02/23/2017] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumour in adults. Current prognosis with standard treatment is poor. Immunotherapy is a new paradigm in tumour management. Specifically, recent advances in the field of immune checkpoint molecules have led to dramatic results in many cancers. Inhibition of one particular, programmed cell death-1 (PD-1) has recently been shown to be highly effective in melanoma and non-small cell lung cancer. There has also been recent data to suggest potential benefit in GBM. There also appears to be a relationship between immune checkpoint inhibition and hypermutation, in particular with the mismatch repair process. In this review we look at the current knowledge of immune checkpoint inhibitors with a focus on the PD-1 pathway. We will also review the evidence of PD-1 inhibition in GBM and the role of hypermutation in PD-1 inhibition.
Collapse
|
194
|
Gymrek M. A genomic view of short tandem repeats. Curr Opin Genet Dev 2017; 44:9-16. [PMID: 28213161 DOI: 10.1016/j.gde.2017.01.012] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/30/2017] [Indexed: 12/31/2022]
Abstract
Short tandem repeats (STRs) are some of the fastest mutating loci in the genome. Tools for accurately profiling STRs from high-throughput sequencing data have enabled genome-wide interrogation of more than a million STRs across hundreds of individuals. These catalogs have revealed that STRs are highly multiallelic and may contribute more de novo mutations than any other variant class. Recent studies have leveraged these catalogs to show that STRs play a widespread role in regulating gene expression and other molecular phenotypes. These analyses suggest that STRs are an underappreciated but rich reservoir of variation that likely make significant contributions to Mendelian diseases, complex traits, and cancer.
Collapse
Affiliation(s)
- Melissa Gymrek
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
195
|
Kautto EA, Bonneville R, Miya J, Yu L, Krook MA, Reeser JW, Roychowdhury S. Performance evaluation for rapid detection of pan-cancer microsatellite instability with MANTIS. Oncotarget 2017; 8:7452-7463. [PMID: 27980218 PMCID: PMC5352334 DOI: 10.18632/oncotarget.13918] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 12/02/2016] [Indexed: 12/28/2022] Open
Abstract
In current clinical practice, microsatellite instability (MSI) and mismatch repair deficiency detection is performed with MSI-PCR and immunohistochemistry. Recent research has produced several computational tools for MSI detection with next-generation sequencing (NGS) data; however a comprehensive analysis of computational methods has not yet been performed. In this study, we introduce a new MSI detection tool, MANTIS, and demonstrate its favorable performance compared to the previously published tools mSINGS and MSISensor. We evaluated 458 normal-tumor sample pairs across six cancer subtypes, testing classification performance on variable numbers of target loci ranging from 10 to 2539. All three computational methods were found to be accurate, with MANTIS exhibiting the highest accuracy with 98.91% of samples from all six diseases classified correctly. MANTIS displayed superior performance among the three tools, having the highest overall sensitivity (MANTIS 97.18%, MSISensor 96.48%, mSINGS 76.06%) and specificity (MANTIS 99.68%, mSINGS 99.68%, MSISensor 98.73%) across six cancer types, even with loci panels of varying size. Additionally, MANTIS also had the lowest resource consumption (<1% of the space and <7% of the memory required by mSINGS) and fastest running times (49.6% and 8.7% of the running times of MSISensor and mSINGS, respectively). This study highlights the potential utility of MANTIS in classifying samples by MSI-status, allowing its incorporation into existing NGS pipelines.
Collapse
Affiliation(s)
- Esko A. Kautto
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Russell Bonneville
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Jharna Miya
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Lianbo Yu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Melanie A. Krook
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Julie W. Reeser
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Sameek Roychowdhury
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
196
|
Abstract
Inflammation affects all stages of tumorigenesis. A key signaling pathway leading to acute and chronic inflammation is through activation of the caspase-1 inflammasome. Inflammasome complexes are assembled on activation of certain nucleotide-binding domain, leucine-rich repeat-containing proteins (NLR), AIM2-like receptors, or pyrin. Of these, NLRP1, NLRP3, NLRC4, NLRP6, and AIM2 influence the pathogenesis of cancer by modulating innate and adaptive immune responses, cell death, proliferation, and/or the gut microbiota. Activation of the inflammasome and IL18 signaling pathways is largely protective in colitis-associated colorectal cancer, whereas excessive inflammation driven by the inflammasome or the IL1 signaling pathways promotes breast cancer, fibrosarcoma, gastric carcinoma, and lung metastasis in a context-dependent manner. The clinical relevance of inflammasomes in multiple forms of cancer highlights their therapeutic promise as molecular targets. In this review, we explore the crossroads between inflammasomes and the development of various tumors and discuss possible therapeutic values in targeting the inflammasome for the prevention and treatment of cancer. Cancer Immunol Res; 5(2); 94-99. ©2017 AACR.
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Si Ming Man
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | |
Collapse
|
197
|
Imielinski M, Guo G, Meyerson M. Insertions and Deletions Target Lineage-Defining Genes in Human Cancers. Cell 2017; 168:460-472.e14. [PMID: 28089356 DOI: 10.1016/j.cell.2016.12.025] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/25/2016] [Accepted: 12/16/2016] [Indexed: 01/21/2023]
Abstract
Certain cell types function as factories, secreting large quantities of one or more proteins that are central to the physiology of the respective organ. Examples include surfactant proteins in lung alveoli, albumin in liver parenchyma, and lipase in the stomach lining. Whole-genome sequencing analysis of lung adenocarcinomas revealed noncoding somatic mutational hotspots near VMP1/MIR21 and indel hotspots in surfactant protein genes (SFTPA1, SFTPB, and SFTPC). Extrapolation to other solid cancers demonstrated highly recurrent and tumor-type-specific indel hotspots targeting the noncoding regions of highly expressed genes defining certain secretory cellular lineages: albumin (ALB) in liver carcinoma, gastric lipase (LIPF) in stomach carcinoma, and thyroglobulin (TG) in thyroid carcinoma. The sequence contexts of indels targeting lineage-defining genes were significantly enriched in the AATAATD DNA motif and specific chromatin contexts, including H3K27ac and H3K36me3. Our findings illuminate a prevalent and hitherto unrecognized mutational process linking cellular lineage and cancer.
Collapse
Affiliation(s)
- Marcin Imielinski
- Department of Pathology and Laboratory Medicine, Englander Institute for Precision Medicine, Institute for Computational Biomedicine, and Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY 10013, USA.
| | - Guangwu Guo
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Matthew Meyerson
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
198
|
Le Gallo M, Lozy F, Bell DW. Next-Generation Sequencing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:119-148. [DOI: 10.1007/978-3-319-43139-0_5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
199
|
Stelloo E, Jansen A, Osse E, Nout R, Creutzberg C, Ruano D, Church D, Morreau H, Smit V, van Wezel T, Bosse T. Practical guidance for mismatch repair-deficiency testing in endometrial cancer. Ann Oncol 2017; 28:96-102. [DOI: 10.1093/annonc/mdw542] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
200
|
Nagahashi M, Wakai T, Shimada Y, Ichikawa H, Kameyama H, Kobayashi T, Sakata J, Yagi R, Sato N, Kitagawa Y, Uetake H, Yoshida K, Oki E, Kudo SE, Izutsu H, Kodama K, Nakada M, Tse J, Russell M, Heyer J, Powers W, Sun R, Ring JE, Takabe K, Protopopov A, Ling Y, Okuda S, Lyle S. Genomic landscape of colorectal cancer in Japan: clinical implications of comprehensive genomic sequencing for precision medicine. Genome Med 2016; 8:136. [PMID: 28007036 PMCID: PMC5180401 DOI: 10.1186/s13073-016-0387-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022] Open
Abstract
Background Comprehensive genomic sequencing (CGS) has the potential to revolutionize precision medicine for cancer patients across the globe. However, to date large-scale genomic sequencing of cancer patients has been limited to Western populations. In order to understand possible ethnic and geographic differences and to explore the broader application of CGS to other populations, we sequenced a panel of 415 important cancer genes to characterize clinically actionable genomic driver events in 201 Japanese patients with colorectal cancer (CRC). Methods Using next-generation sequencing methods, we examined all exons of 415 known cancer genes in Japanese CRC patients (n = 201) and evaluated for concordance among independent data obtained from US patients with CRC (n = 108) and from The Cancer Genome Atlas-CRC whole exome sequencing (WES) database (n = 224). Mutation data from non-hypermutated Japanese CRC patients were extracted and clustered by gene mutation patterns. Two different sets of genes from the 415-gene panel were used for clustering: 61 genes with frequent alteration in CRC and 26 genes that are clinically actionable in CRC. Results The 415-gene panel is able to identify all of the critical mutations in tumor samples as well as WES, including identifying hypermutated tumors. Although the overall mutation spectrum of the Japanese patients is similar to that of the Western population, we found significant differences in the frequencies of mutations in ERBB2 and BRAF. We show that the 415-gene panel identifies a number of clinically actionable mutations in KRAS, NRAS, and BRAF that are not detected by hot-spot testing. We also discovered that 26% of cases have mutations in genes involved in DNA double-strand break repair pathway. Unsupervised clustering revealed that a panel of 26 genes can be used to classify the patients into eight different categories, each of which can optimally be treated with a particular combination therapy. Conclusions Use of a panel of 415 genes can reliably identify all of the critical mutations in CRC patients and this information of CGS can be used to determine the most optimal treatment for patients of all ethnicities. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0387-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hitoshi Kameyama
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Takashi Kobayashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Jun Sakata
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Ryoma Yagi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Nobuaki Sato
- Niigata Cancer Center Hospital, 15-3 Kawagishi-cho 2-Chome, Chuo-ku, Niigata City, Niigata, 951-8566, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35 Shinano-machi, Shinjyuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Uetake
- Department of Chemotherapy and Oncosurgery, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Gifu University School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shin-Ei Kudo
- Digestive Disease Center, Showa University Northern Yokohama Hospital, 35-1 Chigasaki-chuo, Tsuzuki-ku, Yokohama, 224-8503, Japan
| | - Hiroshi Izutsu
- Diagnostics Research Department, Life innovation Research Institute, Denka innovation center, Denka Co., Ltd., 3-5-1 Asahi-Machi, Machida-City, Tokyo, 194-8560, Japan
| | - Keisuke Kodama
- Diagnostics Research Department, Life innovation Research Institute, Denka innovation center, Denka Co., Ltd., 3-5-1 Asahi-Machi, Machida-City, Tokyo, 194-8560, Japan
| | - Mitsutaka Nakada
- Diagnostics Research Department, Life innovation Research Institute, Denka innovation center, Denka Co., Ltd., 3-5-1 Asahi-Machi, Machida-City, Tokyo, 194-8560, Japan
| | - Julie Tse
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA
| | - Meaghan Russell
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA
| | - Joerg Heyer
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA
| | - Winslow Powers
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA
| | - Ruobai Sun
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA
| | - Jennifer E Ring
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA
| | - Kazuaki Takabe
- Breast Surgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY, 14263, USA.,Department of Surgery, University at Buffalo, The State University of New York, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | | | - Yiwei Ling
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | - Stephen Lyle
- KEW, Inc, 840 Memorial Drive, 4th floor, Cambridge, MA, 02139, USA. .,University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| |
Collapse
|