1
|
Kus ME, Sahin C, Kilic E, Askin A, Ozgur MM, Karahanogullari G, Aksit A, O'Connell RM, Ekiz HA. TCGEx: a powerful visual interface for exploring and analyzing cancer gene expression data. EMBO Rep 2025; 26:1863-1890. [PMID: 40033050 PMCID: PMC11976970 DOI: 10.1038/s44319-025-00407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Analyzing gene expression data from the Cancer Genome Atlas (TCGA) and similar repositories often requires advanced coding skills, creating a barrier for many researchers. To address this challenge, we developed The Cancer Genome Explorer (TCGEx), a user-friendly, web-based platform for conducting sophisticated analyses such as survival modeling, gene set enrichment analysis, unsupervised clustering, and linear regression-based machine learning. TCGEx provides access to preprocessed TCGA data and immune checkpoint inhibition studies while allowing integration of user-uploaded data sets. Using TCGEx, we explore molecular subsets of human melanoma and identify microRNAs associated with intratumoral immunity. These findings are validated with independent clinical trial data on immune checkpoint inhibitors for melanoma and other cancers. In addition, we identify cytokine genes that can be used to predict treatment responses to various immune checkpoint inhibitors prior to treatment. Built on the R/Shiny framework, TCGEx offers customizable features to adapt analyses for diverse research contexts and generate publication-ready visualizations. TCGEx is freely available at https://tcgex.iyte.edu.tr , providing an accessible tool to extract insights from cancer transcriptomics data.
Collapse
Affiliation(s)
- M Emre Kus
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Cagatay Sahin
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Emre Kilic
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Arda Askin
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - M Mert Ozgur
- The Department of Molecular Biology and Genetics, Bilkent University, 06800, Cankaya, Ankara, Turkey
| | - Gokhan Karahanogullari
- The Department of Mathematics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Ahmet Aksit
- The Department of Information Technologies, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Ryan M O'Connell
- The Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - H Atakan Ekiz
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey.
| |
Collapse
|
2
|
Kuras M, Betancourt LH, Hong R, Szadai L, Rodriguez J, Horvatovich P, Pla I, Eriksson J, Szeitz B, Deszcz B, Welinder C, Sugihara Y, Ekedahl H, Baldetorp B, Ingvar C, Lundgren L, Lindberg H, Oskolas H, Horvath Z, Rezeli M, Gil J, Appelqvist R, Kemény LV, Malm J, Sanchez A, Szasz AM, Pawłowski K, Wieslander E, Fenyö D, Nemeth IB, Marko-Varga G. Proteogenomic Profiling of Treatment-Naïve Metastatic Malignant Melanoma. Cancers (Basel) 2025; 17:832. [PMID: 40075679 PMCID: PMC11899103 DOI: 10.3390/cancers17050832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Melanoma is a highly heterogeneous disease, and a deeper molecular classification is essential for improving patient stratification and treatment approaches. Here, we describe the histopathology-driven proteogenomic landscape of 142 treatment-naïve metastatic melanoma samples to uncover molecular subtypes and clinically relevant biomarkers. METHODS We performed an integrative proteogenomic analysis to identify proteomic subtypes, assess the impact of BRAF V600 mutations, and study the molecular profiles and cellular composition of the tumor microenvironment. Clinical and histopathological data were used to support findings related to tissue morphology, disease progression, and patient outcomes. RESULTS Our analysis revealed five distinct proteomic subtypes that integrate immune and stromal microenvironment components and correlate with clinical and histopathological parameters. We demonstrated that BRAF V600-mutated melanomas exhibit biological heterogeneity, where an oncogene-induced senescence-like phenotype is associated with improved survival. This led to a proposed mortality risk-based stratification that may contribute to more personalized treatment strategies. Furthermore, tumor microenvironment composition strongly correlated with disease progression and patient outcomes, highlighting a histopathological connective tissue-to-tumor ratio assessment as a potential decision-making tool. We identified a melanoma-associated SAAV signature linked to extracellular matrix remodeling and SAAV-derived neoantigens as potential targets for anti-tumor immune responses. CONCLUSIONS This study provides a comprehensive stratification of metastatic melanoma, integrating proteogenomic insights with histopathological features. The findings may aid in the development of tailored diagnostic and therapeutic strategies, improving patient management and outcomes.
Collapse
Affiliation(s)
- Magdalena Kuras
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Lazaro Hiram Betancourt
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Runyu Hong
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; (R.H.); (D.F.)
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Leticia Szadai
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.S.); (I.B.N.)
| | - Jimmy Rodriguez
- Department of Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden;
| | - Peter Horvatovich
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
- Department of Analytical Biochemistry, Faculty of Science and Engineering, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Indira Pla
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Jonatan Eriksson
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1085 Budapest, Hungary
| | - Bartłomiej Deszcz
- Department of Biochemistry and Microbiology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Charlotte Welinder
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Yutaka Sugihara
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Henrik Ekedahl
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
- SUS University Hospital Lund, 222 42 Lund, Sweden;
| | - Bo Baldetorp
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Christian Ingvar
- SUS University Hospital Lund, 222 42 Lund, Sweden;
- Department of Surgery, Clinical Sciences, Lund University, SUS, 221 00 Lund, Sweden
| | - Lotta Lundgren
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
- SUS University Hospital Lund, 222 42 Lund, Sweden;
| | - Henrik Lindberg
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Henriett Oskolas
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Zsolt Horvath
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Jeovanis Gil
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | - Roger Appelqvist
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Lajos V. Kemény
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1085 Budapest, Hungary;
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Lendület “Momentum” Dermatology Research Group, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| | - Johan Malm
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | - Aniel Sanchez
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | | | - Krzysztof Pawłowski
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
- Department of Biochemistry and Microbiology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elisabet Wieslander
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; (R.H.); (D.F.)
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Istvan Balazs Nemeth
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.S.); (I.B.N.)
| | - György Marko-Varga
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- 1st Department of Surgery, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
3
|
Besedina E, Supek F. Copy number losses of oncogenes and gains of tumor suppressor genes generate common driver mutations. Nat Commun 2024; 15:6139. [PMID: 39033140 PMCID: PMC11271286 DOI: 10.1038/s41467-024-50552-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 07/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cancer driver genes can undergo positive selection for various types of genetic alterations, including gain-of-function or loss-of-function mutations and copy number alterations (CNA). We investigated the landscape of different types of alterations affecting driver genes in 17,644 cancer exomes and genomes. We find that oncogenes may simultaneously exhibit signatures of positive selection and also negative selection in different gene segments, suggesting a method to identify additional tumor types where an oncogene is a driver or a vulnerability. Next, we characterize the landscape of CNA-dependent selection effects, revealing a general trend of increased positive selection on oncogene mutations not only upon CNA gains but also upon CNA deletions. Similarly, we observe a positive interaction between mutations and CNA gains in tumor suppressor genes. Thus, two-hit events involving point mutations and CNA are universally observed regardless of the type of CNA and may signal new therapeutic opportunities. An analysis with focus on the somatic CNA two-hit events can help identify additional driver genes relevant to a tumor type. By a global inference of point mutation and CNA selection signatures and interactions thereof across genes and tissues, we identify 9 evolutionary archetypes of driver genes, representing different mechanisms of (in)activation by genetic alterations.
Collapse
Affiliation(s)
- Elizaveta Besedina
- Institute for Research in Biomedicine (IRB Barcelona), 08028, Barcelona, Spain
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), 08028, Barcelona, Spain.
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, 2200, Copenhagen, Denmark.
- Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain.
| |
Collapse
|
4
|
Song S, Koh Y, Kim S, Lee SM, Kim HU, Ko JM, Lee SH, Yoon SS, Park S. Systematic analysis of Mendelian disease-associated gene variants reveals new classes of cancer-predisposing genes. Genome Med 2023; 15:107. [PMID: 38143269 PMCID: PMC10749499 DOI: 10.1186/s13073-023-01252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/30/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND Despite the acceleration of somatic driver gene discovery facilitated by recent large-scale tumor sequencing data, the contribution of inherited variants remains largely unexplored, primarily focusing on previously known cancer predisposition genes (CPGs) due to the low statistical power associated with detecting rare pathogenic variant-phenotype associations. METHODS Here, we introduce a generalized log-regression model to measure the excess of pathogenic variants within genes in cancer patients compared to control samples. It aims to measure gene-level cancer risk enrichment by collapsing rare pathogenic variants after controlling the population differences across samples. RESULTS In this study, we investigate whether pathogenic variants in Mendelian disease-associated genes (OMIM genes) are enriched in cancer patients compared to controls. Utilizing data from PCAWG and the 1,000 Genomes Project, we identify 103 OMIM genes demonstrating significant enrichment of pathogenic variants in cancer samples (FDR 20%). Through an integrative approach considering three distinct properties, we classify these CPG-like OMIM genes into four clusters, indicating potential diverse mechanisms underlying tumor progression. Further, we explore the function of PAH (a key metabolic enzyme associated with Phenylketonuria), the gene exhibiting the highest prevalence of pathogenic variants in a pan-cancer (1.8%) compared to controls (0.6%). CONCLUSIONS Our findings suggest a possible cancer progression mechanism through metabolic profile alterations. Overall, our data indicates that pathogenic OMIM gene variants contribute to cancer progression and introduces new CPG classifications potentially underpinning diverse tumorigenesis mechanisms.
Collapse
Affiliation(s)
- Seulki Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Structural Biology Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Calle de Melchor Fernández Almagro, 3, Madrid, 28029, Spain
| | - Youngil Koh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Biomedical Research Institute and Departments of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Seokhyeon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Sang Mi Lee
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyun Uk Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| | - Jung Min Ko
- Department of Pediatrics, Seoul National University Children's Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea.
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Biomedical Research Institute and Departments of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Republic of Korea.
| | - Solip Park
- Structural Biology Program, Centro Nacional de Investigaciones Oncológicas (CNIO), Calle de Melchor Fernández Almagro, 3, Madrid, 28029, Spain.
| |
Collapse
|
5
|
Chen C, Lin CJ, Pei YC, Ma D, Liao L, Li SY, Fan L, Di GH, Wu SY, Liu XY, Wang YJ, Hong Q, Zhang GL, Xu LL, Li BB, Huang W, Shi JX, Jiang YZ, Hu X, Shao ZM. Comprehensive genomic profiling of breast cancers characterizes germline-somatic mutation interactions mediating therapeutic vulnerabilities. Cell Discov 2023; 9:125. [PMID: 38114467 PMCID: PMC10730692 DOI: 10.1038/s41421-023-00614-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/08/2023] [Indexed: 12/21/2023] Open
Abstract
Germline-somatic mutation interactions are universal and associated with tumorigenesis, but their role in breast cancer, especially in non-Caucasians, remains poorly characterized. We performed large-scale prospective targeted sequencing of matched tumor-blood samples from 4079 Chinese females, coupled with detailed clinical annotation, to map interactions between germline and somatic alterations. We discovered 368 pathogenic germline variants and identified 5 breast cancer DNA repair-associated genes (BCDGs; BRCA1/BRCA2/CHEK2/PALB2/TP53). BCDG mutation carriers, especially those with two-hit inactivation, demonstrated younger onset, higher tumor mutation burden, and greater clinical benefits from platinum drugs, PARP inhibitors, and immune checkpoint inhibitors. Furthermore, we leveraged a multiomics cohort to reveal that clinical benefits derived from two-hit events are associated with increased genome instability and an immune-activated tumor microenvironment. We also established an ethnicity-specific tool to predict BCDG mutation and two-hit status for genetic evaluation and therapeutic decisions. Overall, this study leveraged the large sequencing cohort of Chinese breast cancers, optimizing genomics-guided selection of DNA damaging-targeted therapy and immunotherapy within a broader population.
Collapse
Affiliation(s)
- Chao Chen
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cai-Jin Lin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Chen Pei
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ding Ma
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Li Liao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Si-Yuan Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Fan
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gen-Hong Di
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Song-Yang Wu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi-Yu Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun-Jin Wang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qi Hong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guo-Liang Zhang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lin-Lin Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bei-Bei Li
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wei Huang
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jin-Xiu Shi
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Xin Hu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Precision Cancer Medical Center Affiliated to Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
6
|
Perdomo-González DI, Id-Lahoucine S, Molina A, Cánovas A, Laseca N, Azor PJ, Valera M. Transmission ratio distortion detection by neutral genetic markers in the Pura Raza Española horse breed. Animal 2023; 17:101012. [PMID: 37950978 DOI: 10.1016/j.animal.2023.101012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 11/13/2023] Open
Abstract
Transmission Ratio Distortion (TRD) is a genetic phenomenon widely demonstrated in several livestock species, but barely in equine species. The TRD occurs when certain genotypes are over- or under-represented in the offspring of a particular mating and can be caused by a variety of factors during gamete formation or during embryonic development. For this study, 126 394 trios consisting of a stallion, mare, and offspring were genotyped using a panel of 17 neutral microsatellite markers recommended by the International Society for Animal Genetics for paternity tests and individual identification. The number of alleles available for each marker ranges from 13 to 18, been 268 the total number of alleles investigated. The TRDscan v.2.0 software was used with the biallelic procedure to identify regions with distorted segregation ratios. After completing the analysis, a total of 12 alleles (out of 11 microsatellites) were identified with decisive evidence for genotypic TRD; 3 and 9 with additive and heterosis patterns, respectively. In addition, 19 alleles (out of 10 microsatellites) were identified displaying allelic TRD. Among them, 14 and 5 were parent-unspecific and stallion-mare-specific TRD. Out of the TRD regions, 24 genes were identified and annotated, predominantly associated with cholesterol metabolism and homeostasis. These genes are often linked to non-specific symptoms like impaired fertility, stunted growth, and compromised overall health. The results suggest a significant impact on the inheritance of certain genetic traits in horses. Further analysis and validation are needed to better understand the TRD impact before the potential implementation in the horse breeding programme strategies.
Collapse
Affiliation(s)
| | - S Id-Lahoucine
- Department of Animal and Veterinary Science, Scotland's Rural College, Easter Bush, Edinburgh EH25 9RG, United Kingdom
| | - A Molina
- Departamento de Genética, Universidad de Córdoba, Córdoba 14014, Spain
| | - A Cánovas
- Center of Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - N Laseca
- Departamento de Genética, Universidad de Córdoba, Córdoba 14014, Spain
| | - P J Azor
- Real Asociación Nacional de Criadores de Caballos de Pura Raza Española (ANCCE), Sevilla 41014, Spain
| | - M Valera
- Departamento de Agronomía, ETSIA, Universidad de Sevilla, Sevilla 41005, Spain
| |
Collapse
|
7
|
Rane JK, Frankell AM, Weeden CE, Swanton C. Clonal Evolution in Healthy and Premalignant Tissues: Implications for Early Cancer Interception Strategies. Cancer Prev Res (Phila) 2023; 16:369-378. [PMID: 36930945 PMCID: PMC7614725 DOI: 10.1158/1940-6207.capr-22-0469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/17/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
Histologically normal human tissues accumulate significant mutational burden with age. The extent and spectra of mutagenesis are comparable both in rapidly proliferating and post-mitotic tissues and in stem cells compared with their differentiated progeny. Some of these mutations provide increased fitness, giving rise to clones which, at times, can replace the entire surface area of tissues. Compared with cancer, somatic mutations in histologically normal tissues are primarily single-nucleotide variations. Interestingly though, the presence of these mutations and positive clonal selection in isolation remains a poor indicator of potential future cancer transformation in solid tissues. Common clonally expanded mutations in histologically normal tissues also do not always represent the most frequent early mutations in cancers of corresponding tissues, indicating differences in selection pressures. Preliminary evidence implies that stroma and immune system co-evolve with age, which may impact selection dynamics. In this review, we will explore the mutational landscape of histologically normal and premalignant human somatic tissues in detail and discuss cell-intrinsic and environmental factors that can determine the fate of positively selected mutations within them. Precisely pinpointing these determinants of cancer transformation would aid development of early cancer interventional and prevention strategies.
Collapse
Affiliation(s)
- Jayant K. Rane
- University College London Cancer Institute, London, UK
- Department of Clinical Oncology, University College London Hospitals, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Alexander M. Frankell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare E. Weeden
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Medical Oncology, University College London Hospitals, London, UK
| |
Collapse
|
8
|
Casasent AK, Almekinders MM, Mulder C, Bhattacharjee P, Collyar D, Thompson AM, Jonkers J, Lips EH, van Rheenen J, Hwang ES, Nik-Zainal S, Navin NE, Wesseling J. Learning to distinguish progressive and non-progressive ductal carcinoma in situ. Nat Rev Cancer 2022; 22:663-678. [PMID: 36261705 DOI: 10.1038/s41568-022-00512-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 02/07/2023]
Abstract
Ductal carcinoma in situ (DCIS) is a non-invasive breast neoplasia that accounts for 25% of all screen-detected breast cancers diagnosed annually. Neoplastic cells in DCIS are confined to the ductal system of the breast, although they can escape and progress to invasive breast cancer in a subset of patients. A key concern of DCIS is overtreatment, as most patients screened for DCIS and in whom DCIS is diagnosed will not go on to exhibit symptoms or die of breast cancer, even if left untreated. However, differentiating low-risk, indolent DCIS from potentially progressive DCIS remains challenging. In this Review, we summarize our current knowledge of DCIS and explore open questions about the basic biology of DCIS, including those regarding how genomic events in neoplastic cells and the surrounding microenvironment contribute to the progression of DCIS to invasive breast cancer. Further, we discuss what information will be needed to prevent overtreatment of indolent DCIS lesions without compromising adequate treatment for high-risk patients.
Collapse
Affiliation(s)
- Anna K Casasent
- Department of Genetics, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Charlotta Mulder
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | | | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Esther H Lips
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Serena Nik-Zainal
- Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Nicholas E Navin
- Department of Genetics, MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioinformatics, MD Anderson Cancer Center, Houston, TX, USA
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands.
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
9
|
Vali-Pour M, Park S, Espinosa-Carrasco J, Ortiz-Martínez D, Lehner B, Supek F. The impact of rare germline variants on human somatic mutation processes. Nat Commun 2022; 13:3724. [PMID: 35764656 PMCID: PMC9240060 DOI: 10.1038/s41467-022-31483-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/17/2022] [Indexed: 02/07/2023] Open
Abstract
Somatic mutations are an inevitable component of ageing and the most important cause of cancer. The rates and types of somatic mutation vary across individuals, but relatively few inherited influences on mutation processes are known. We perform a gene-based rare variant association study with diverse mutational processes, using human cancer genomes from over 11,000 individuals of European ancestry. By combining burden and variance tests, we identify 207 associations involving 15 somatic mutational phenotypes and 42 genes that replicated in an independent data set at a false discovery rate of 1%. We associate rare inherited deleterious variants in genes such as MSH3, EXO1, SETD2, and MTOR with two phenotypically different forms of DNA mismatch repair deficiency, and variants in genes such as EXO1, PAXIP1, RIF1, and WRN with deficiency in homologous recombination repair. In addition, we identify associations with other mutational processes, such as APEX1 with APOBEC-signature mutagenesis. Many of the genes interact with each other and with known mutator genes within cellular sub-networks. Considered collectively, damaging variants in the identified genes are prevalent in the population. We suggest that rare germline variation in diverse genes commonly impacts mutational processes in somatic cells.
Collapse
Affiliation(s)
- Mischan Vali-Pour
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Solip Park
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Jose Espinosa-Carrasco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Daniel Ortiz-Martínez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Ben Lehner
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| |
Collapse
|
10
|
Levatić J, Salvadores M, Fuster-Tormo F, Supek F. Mutational signatures are markers of drug sensitivity of cancer cells. Nat Commun 2022; 13:2926. [PMID: 35614096 PMCID: PMC9132939 DOI: 10.1038/s41467-022-30582-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/09/2022] [Indexed: 02/06/2023] Open
Abstract
Genomic analyses have revealed mutational footprints associated with DNA maintenance gone awry, or with mutagen exposures. Because cancer therapeutics often target DNA synthesis or repair, we asked if mutational signatures make useful markers of drug sensitivity. We detect mutational signatures in cancer cell line exomes (where matched healthy tissues are not available) by adjusting for the confounding germline mutation spectra across ancestries. We identify robust associations between various mutational signatures and drug activity across cancer cell lines; these are as numerous as associations with established genetic markers such as driver gene alterations. Signatures of prior exposures to DNA damaging agents - including chemotherapy - tend to associate with drug resistance, while signatures of deficiencies in DNA repair tend to predict sensitivity towards particular therapeutics. Replication analyses across independent drug and CRISPR genetic screening data sets reveal hundreds of robust associations, which are provided as a resource for drug repurposing guided by mutational signature markers.
Collapse
Affiliation(s)
- Jurica Levatić
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, C/ Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Marina Salvadores
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, C/ Baldiri Reixac 10, 08028, Barcelona, Spain
| | - Francisco Fuster-Tormo
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, C/ Baldiri Reixac 10, 08028, Barcelona, Spain
- MDS Group, Josep Carreras Leukaemia Research Institute, Ctra de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Spain
| | - Fran Supek
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, C/ Baldiri Reixac 10, 08028, Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig de Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
11
|
Gómez-Flores-Ramos L, Barraza-Arellano AL, Mohar A, Trujillo-Martínez M, Grimaldo L, Ortiz-Lopez R, Treviño V. Germline Variants in Cancer Genes from Young Breast Cancer Mexican Patients. Cancers (Basel) 2022; 14:cancers14071647. [PMID: 35406420 PMCID: PMC8997148 DOI: 10.3390/cancers14071647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/03/2022] [Accepted: 03/18/2022] [Indexed: 02/05/2023] Open
Abstract
Breast cancer (BC) is one of the most frequent cancer types in women worldwide. About 7% is diagnosed in young women (YBC) less than 40 years old. In Mexico, however, YBC reaches 15% suggesting a higher genetic susceptibility. There have been some reports of germline variants in YBC across the world. However, there is only one report from a Mexican population, which is not restricted by age and limited to a panel of 143 genes resulting in 15% of patients carrying putatively pathogenic variants. Nevertheless, expanding the analysis to whole exome involves using more complex tools to determine which genes and variants could be pathogenic. We used germline whole exome sequencing combined with the PeCanPie tool to analyze exome variants in 115 YBC patients. Our results showed that we were able to identify 49 high likely pathogenic variants involving 40 genes on 34% of patients. We noted many genes already reported in BC and YBC worldwide, such as BRCA1, BRCA2, ATM, CHEK2, PALB2, and POLQ, but also others not commonly reported in YBC in Latin America, such as CLTCL1, DDX3X, ERCC6, FANCE, and NFKBIE. We show further supporting and controversial evidence for some of these genes. We conclude that exome sequencing combined with robust annotation tools and further analysis, can identify more genes and more patients affected by germline mutations in cancer.
Collapse
Affiliation(s)
- Liliana Gómez-Flores-Ramos
- CONACYT/Center for Population Health Research, National Institute of Public Health, Universidad No. 655, Cuernavaca 62100, Morelos, Mexico; (L.G.-F.-R.); (L.G.)
| | - Angélica Leticia Barraza-Arellano
- School of Medicine, Tecnologico de Monterrey, Morones Prieto Av 3000, Los Doctores, Monterrey 64710, Nuevo Leon, Mexico; (A.L.B.-A.); (R.O.-L.)
| | - Alejandro Mohar
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Dirección de Investigación, Instituto Nacional de Cancerología, Av. San Fernando #22, Col. Sección XVI, Delegación Tlalpan, Mexico City 14080, Mexico;
| | - Miguel Trujillo-Martínez
- Instituto Mexicano del Seguro Social, Hospital General de Zona con Medicina Familiar No. 7, Cuautla 62780, Morelos, Mexico;
| | - Lizbeth Grimaldo
- CONACYT/Center for Population Health Research, National Institute of Public Health, Universidad No. 655, Cuernavaca 62100, Morelos, Mexico; (L.G.-F.-R.); (L.G.)
| | - Rocío Ortiz-Lopez
- School of Medicine, Tecnologico de Monterrey, Morones Prieto Av 3000, Los Doctores, Monterrey 64710, Nuevo Leon, Mexico; (A.L.B.-A.); (R.O.-L.)
- The Institute for Obesity Research, Tecnologico de Monterrey, Eugenio Garza Sada Av 2501, Monterrey 64849, Nuevo Leon, Mexico
| | - Víctor Treviño
- School of Medicine, Tecnologico de Monterrey, Morones Prieto Av 3000, Los Doctores, Monterrey 64710, Nuevo Leon, Mexico; (A.L.B.-A.); (R.O.-L.)
- The Institute for Obesity Research, Tecnologico de Monterrey, Eugenio Garza Sada Av 2501, Monterrey 64849, Nuevo Leon, Mexico
- Correspondence:
| |
Collapse
|
12
|
Stålhammar G, Yeung A, Mendoza P, Dubovy SR, William Harbour J, Grossniklaus HE. Gain of Chromosome 6p Correlates with Severe Anaplasia, Cellular Hyperchromasia, and Extraocular Spread of Retinoblastoma. OPHTHALMOLOGY SCIENCE 2022; 2:100089. [PMID: 36246172 PMCID: PMC9560556 DOI: 10.1016/j.xops.2021.100089] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/03/2021] [Accepted: 12/03/2021] [Indexed: 06/16/2023]
Abstract
PURPOSE Gain of chromosome 6p has been associated with poor ocular survival in retinoblastoma and histopathologic grading of anaplasia with increased risk of metastatic spread and death. This study examined the correlation between these factors and other chromosomal abnormalities as well as results of whole genome sequencing, digital morphometry, and progression-free survival. DESIGN Retrospective cohort study from 2 United States tertiary referral centers. PARTICIPANTS Forty-two children who had undergone enucleation for retinoblastoma from January 2000 through December 2017. METHODS Status of chromosomes 6p, 1q, 9q, and 16q was evaluated with fluorescence in situ hybridization, the degree of anaplasia and presence of histologic high-risk features were assessed by ocular pathologists, digital morphometry was performed on scanned tumor slides, and whole genome sequencing was performed on a subset of tumors. Progression-free survival was defined as absence of distant or local metastases or tumor growth beyond the cut end of the optic nerve. MAIN OUTCOME MEASURES Correlation between each of chromosomal abnormalities, anaplasia, morphometry and sequencing results, and survival. RESULTS Forty-one of 42 included patients underwent primary enucleation and 1 was treated first with intra-arterial chemotherapy. Seven tumors showed mild anaplasia, 19 showed moderate anaplasia, and 16 showed severe anaplasia. All tumors had gain of 1q, 18 tumors had gain of 6p, 6 tumors had gain of 9q, and 36 tumors had loss of 16q. Tumors with severe anaplasia were significantly more likely to harbor 6p gains than tumors with nonsevere anaplasia (P < 0.001). Further, the hematoxylin staining intensity was significantly greater and that of eosin staining significantly lower in tumors with severe anaplasia (P < 0.05). Neither severe anaplasia (P = 0.10) nor gain of 6p (P = 0.21) correlated with histologic high-risk features, and severe anaplasia did not correlate to RB1, CREBBP, NSD1, or BCOR mutations in a subset of 14 tumors (P > 0.5). Patients with gain of 6p showed significantly shorter progression-free survival (P = 0.03, Wilcoxon test). CONCLUSIONS Gain of chromosome 6p emerges as a strong prognostic biomarker in retinoblastoma because it correlates with severe anaplasia, quantifiable changes in tumor cell staining characteristics, and extraocular spread.
Collapse
Affiliation(s)
- Gustav Stålhammar
- Ocular Pathology Service, St. Erik Eye Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Aaron Yeung
- Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Pia Mendoza
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, Georgia
| | - Sander R. Dubovy
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - J. William Harbour
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Hans E. Grossniklaus
- Departments of Ophthalmology and Pathology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
13
|
Quintana I, Mur P, Terradas M, García-Mulero S, Aiza G, Navarro M, Piñol V, Brunet J, Moreno V, Sanz-Pamplona R, Capellá G, Valle L. Potential Involvement of NSD1, KRT24 and ACACA in the Genetic Predisposition to Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14030699. [PMID: 35158968 PMCID: PMC8833793 DOI: 10.3390/cancers14030699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 01/26/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Methods used for the identification of hereditary cancer genes have evolved in parallel to technological progress; however, much of the genetic predisposition to cancer remains unexplained. A new in silico method based on Knudson’s two-hit hypothesis recently identified ~50 putative cancer predisposing genes, but their actual association with cancer has not yet been validated. In our study, we aimed to assess the involvement of these genes in familial/early-onset colorectal cancer (CRC) using different lines of evidence. Our results indicated that most of those genes were not associated with a genetic predisposition to CRC, but suggested a possible association for NSD1, KRT24 and ACACA. Abstract The ALFRED (Allelic Loss Featuring Rare Damaging) in silico method was developed to identify cancer predisposition genes through the identification of somatic second hits. By applying ALFRED to ~10,000 tumor exomes, 49 candidate genes were identified. We aimed to assess the causal association of the identified genes with colorectal cancer (CRC) predisposition. Of the 49 genes, NSD1, HDAC10, KRT24, ACACA and TP63 were selected based on specific criteria relevant for hereditary CRC genes. Gene sequencing was performed in 736 patients with familial/early onset CRC or polyposis without germline pathogenic variants in known genes. Twelve (predicted) damaging variants in 18 patients were identified. A gene-based burden test in 1596 familial/early-onset CRC patients, 271 polyposis patients, 543 TCGA CRC patients and >134,000 controls (gnomAD, non-cancer), revealed no clear association with CRC for any of the studied genes. Nevertheless, (non-significant) over-representation of disruptive variants in NSD1, KRT24 and ACACA in CRC patients compared to controls was observed. A somatic second hit was identified in one of 20 tumors tested, corresponding to an NSD1 carrier. In conclusion, most genes identified through the ALFRED in silico method were not relevant for CRC predisposition, although a possible association was detected for NSD1, KRT24 and ACACA.
Collapse
Affiliation(s)
- Isabel Quintana
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
| | - Pilar Mur
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Mariona Terradas
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
| | - Sandra García-Mulero
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology, Hospitalet de Llobregat, 08908 Barcelona, Spain; (S.G.-M.); (V.M.); (R.S.-P.)
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Gemma Aiza
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
| | - Matilde Navarro
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Virginia Piñol
- Gastroenterology Unit, Hospital Universitario de Girona Dr. Josep Trueta, 17007 Girona, Spain;
| | - Joan Brunet
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Catalan Institute of Oncology, IDIBGi, 17007 Girona, Spain
| | - Victor Moreno
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology, Hospitalet de Llobregat, 08908 Barcelona, Spain; (S.G.-M.); (V.M.); (R.S.-P.)
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, 08907 Barcelona, Spain
| | - Rebeca Sanz-Pamplona
- Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology, Hospitalet de Llobregat, 08908 Barcelona, Spain; (S.G.-M.); (V.M.); (R.S.-P.)
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Gabriel Capellá
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Laura Valle
- Hereditary Cancer Program, Catalan Institute of Oncology, Oncobell Program, IDIBELL, Hospitalet de Llobregat, 08908 Barcelona, Spain; (I.Q.); (P.M.); (M.T.); (G.A.); (M.N.); (J.B.); (G.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
14
|
Park S, Supek F, Lehner B. Higher order genetic interactions switch cancer genes from two-hit to one-hit drivers. Nat Commun 2021; 12:7051. [PMID: 34862370 PMCID: PMC8642467 DOI: 10.1038/s41467-021-27242-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/09/2021] [Indexed: 11/10/2022] Open
Abstract
The classic two-hit model posits that both alleles of a tumor suppressor gene (TSG) must be inactivated to cause cancer. In contrast, for some oncogenes and haploinsufficient TSGs, a single genetic alteration can suffice to increase tumor fitness. Here, by quantifying the interactions between mutations and copy number alterations (CNAs) across 10,000 tumors, we show that many cancer genes actually switch between acting as one-hit or two-hit drivers. Third order genetic interactions identify the causes of some of these switches in dominance and dosage sensitivity as mutations in other genes in the same biological pathway. The correct genetic model for a gene thus depends on the other mutations in a genome, with a second hit in the same gene or an alteration in a different gene in the same pathway sometimes representing alternative evolutionary paths to cancer.
Collapse
Affiliation(s)
- Solip Park
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Fran Supek
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | - Ben Lehner
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
15
|
Kim M, Park J, Bouhaddou M, Kim K, Rojc A, Modak M, Soucheray M, McGregor MJ, O'Leary P, Wolf D, Stevenson E, Foo TK, Mitchell D, Herrington KA, Muñoz DP, Tutuncuoglu B, Chen KH, Zheng F, Kreisberg JF, Diolaiti ME, Gordan JD, Coppé JP, Swaney DL, Xia B, van 't Veer L, Ashworth A, Ideker T, Krogan NJ. A protein interaction landscape of breast cancer. Science 2021; 374:eabf3066. [PMID: 34591612 PMCID: PMC9040556 DOI: 10.1126/science.abf3066] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Minkyu Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Jisoo Park
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Mehdi Bouhaddou
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kyumin Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Ajda Rojc
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Maya Modak
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Margaret Soucheray
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Michael J McGregor
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Patrick O'Leary
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Denise Wolf
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Erica Stevenson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Tzeh Keong Foo
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Dominique Mitchell
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Kari A Herrington
- Department of Biochemistry and Biophysics, Center for Advanced Light Microscopy, University of California, San Francisco, CA, USA
| | - Denise P Muñoz
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Beril Tutuncuoglu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kuei-Ho Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Fan Zheng
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Jason F Kreisberg
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Morgan E Diolaiti
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - John D Gordan
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Jean-Philippe Coppé
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Danielle L Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Bing Xia
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Laura van 't Veer
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Alan Ashworth
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Trey Ideker
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA.,Department of Bioengineering, University of California, San Diego, CA, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| |
Collapse
|
16
|
Ménard T, Rolo D, Koneswarakantha B. Clinical Quality in Cancer Research: Strategy to Assess Data Integrity of Germline Variants Inferred from Tumor-Only Testing Sequencing Data. Pharmaceut Med 2021; 35:225-233. [PMID: 34436760 DOI: 10.1007/s40290-021-00399-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/31/2022]
Abstract
In the majority of cancers, pathogenic variants are only found at the level of the tumor; however, an unusual number of cancers and/or diagnoses at an early age in a single family may suggest a genetic predisposition. Predisposition plays a major role in about 5-10% of adult cancers and in certain childhood tumors. As access to genomic testing for cancer patients continues to expand, the identification of potential germline pathogenic variants (PGPVs) through tumor-DNA sequencing is also increasing. Statistical methods have been developed to infer the presence of a PGPV without the need of a matched normal sample. These methods are mainly used for exploratory research, for example in real-world clinico-genomic databases/platforms (CGDB). These databases are being developed to support many applications, such as targeted drug development, clinical trial optimization, and postmarketing studies. To ensure the integrity of data used for research, a quality management system should be established, and quality oversight activities should be conducted to assess and mitigate clinical quality risks (for patient safety and data integrity). As opposed to well-defined 'good practice' quality guidelines (GxP) areas such as good clinical practice, there are no comprehensive instructions on how to assess the clinical quality of statistically derived variables from sequencing data such as PGPVs. In this article, we aim to share our strategy and propose a possible set of tactics to assess the PGPV quality and to ensure data integrity in exploratory research.
Collapse
|
17
|
Tauchmann S, Schwaller J. NSD1: A Lysine Methyltransferase between Developmental Disorders and Cancer. Life (Basel) 2021; 11:life11090877. [PMID: 34575025 PMCID: PMC8465848 DOI: 10.3390/life11090877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 01/25/2023] Open
Abstract
Recurrent epigenomic alterations associated with multiple human pathologies have increased the interest in the nuclear receptor binding SET domain protein 1 (NSD1) lysine methyltransferase. Here, we review the current knowledge about the biochemistry, cellular function and role of NSD1 in human diseases. Several studies have shown that NSD1 controls gene expression by methylation of lysine 36 of histone 3 (H3K36me1/2) in a complex crosstalk with de novo DNA methylation. Inactivation in flies and mice revealed that NSD1 is essential for normal development and that it regulates multiple cell type-specific functions by interfering with transcriptional master regulators. In humans, putative loss of function NSD1 mutations characterize developmental syndromes, such as SOTOS, as well as cancer from different organs. In pediatric hematological malignancies, a recurrent chromosomal translocation forms a NUP98-NSD1 fusion with SET-dependent leukemogenic activity, which seems targetable by small molecule inhibitors. To treat or prevent diseases driven by aberrant NSD1 activity, future research will need to pinpoint the mechanistic correlation between the NSD1 gene dosage and/or mutational status with development, homeostasis, and malignant transformation.
Collapse
|
18
|
Zhang L, Shi J, Ouyang J, Zhang R, Tao Y, Yuan D, Lv C, Wang R, Ning B, Roberts R, Tong W, Liu Z, Shi T. X-CNV: genome-wide prediction of the pathogenicity of copy number variations. Genome Med 2021; 13:132. [PMID: 34407882 PMCID: PMC8375180 DOI: 10.1186/s13073-021-00945-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/30/2021] [Indexed: 01/04/2023] Open
Abstract
Background Gene copy number variations (CNVs) contribute to genetic diversity and disease prevalence across populations. Substantial efforts have been made to decipher the relationship between CNVs and pathogenesis but with limited success. Results We have developed a novel computational framework X-CNV (www.unimd.org/XCNV), to predict the pathogenicity of CNVs by integrating more than 30 informative features such as allele frequency (AF), CNV length, CNV type, and some deleterious scores. Notably, over 14 million CNVs across various ethnic groups, covering nearly 93% of the human genome, were unified to calculate the AF. X-CNV, which yielded area under curve (AUC) values of 0.96 and 0.94 in training and validation sets, was demonstrated to outperform other available tools in terms of CNV pathogenicity prediction. A meta-voting prediction (MVP) score was developed to quantitively measure the pathogenic effect, which is based on the probabilistic value generated from the XGBoost algorithm. The proposed MVP score demonstrated a high discriminative power in determining pathogenetic CNVs for inherited traits/diseases in different ethnic groups. Conclusions The ability of the X-CNV framework to quantitatively prioritize functional, deleterious, and disease-causing CNV on a genome-wide basis outperformed current CNV-annotation tools and will have broad utility in population genetics, disease-association studies, and diagnostic screening. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-021-00945-4.
Collapse
Affiliation(s)
- Li Zhang
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China.,School of Statistics, Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, East China Normal University, Shanghai, 200062, China
| | - Jingru Shi
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jian Ouyang
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Riquan Zhang
- School of Statistics, Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, East China Normal University, Shanghai, 200062, China
| | - Yiran Tao
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dongsheng Yuan
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Chengkai Lv
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ruiyuan Wang
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Baitang Ning
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Ruth Roberts
- ApconiX Ltd, Alderley Park, Alderley Edge, SK10 4TG, UK.,University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Weida Tong
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Zhichao Liu
- National Center for Toxicological Research, Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Tieliu Shi
- Center for Bioinformatics and Computational Biology, and the Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China. .,School of Statistics, Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, East China Normal University, Shanghai, 200062, China. .,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing, 100083, China.
| |
Collapse
|
19
|
Brandes N, Linial N, Linial M. Genetic association studies of alterations in protein function expose recessive effects on cancer predisposition. Sci Rep 2021; 11:14901. [PMID: 34290314 PMCID: PMC8295298 DOI: 10.1038/s41598-021-94252-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
The characterization of germline genetic variation affecting cancer risk, known as cancer predisposition, is fundamental to preventive and personalized medicine. Studies of genetic cancer predisposition typically identify significant genomic regions based on family-based cohorts or genome-wide association studies (GWAS). However, the results of such studies rarely provide biological insight or functional interpretation. In this study, we conducted a comprehensive analysis of cancer predisposition in the UK Biobank cohort using a new gene-based method for detecting protein-coding genes that are functionally interpretable. Specifically, we conducted proteome-wide association studies (PWAS) to identify genetic associations mediated by alterations to protein function. With PWAS, we identified 110 significant gene-cancer associations in 70 unique genomic regions across nine cancer types and pan-cancer. In 48 of the 110 PWAS associations (44%), estimated gene damage is associated with reduced rather than elevated cancer risk, suggesting a protective effect. Together with standard GWAS, we implicated 145 unique genomic loci with cancer risk. While most of these genomic regions are supported by external evidence, our results also highlight many novel loci. Based on the capacity of PWAS to detect non-additive genetic effects, we found that 46% of the PWAS-significant cancer regions exhibited exclusive recessive inheritance. These results highlight the importance of recessive genetic effects, without relying on familial studies. Finally, we show that many of the detected genes exert substantial cancer risk in the studied cohort determined by a quantitative functional description, suggesting their relevance for diagnosis and genetic consulting.
Collapse
Affiliation(s)
- Nadav Brandes
- grid.9619.70000 0004 1937 0538The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathan Linial
- grid.9619.70000 0004 1937 0538The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Linial
- grid.9619.70000 0004 1937 0538Department of Biological Chemistry, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
20
|
Choi YY, Shin SJ, Lee JE, Madlensky L, Lee ST, Park JS, Jo JH, Kim H, Nachmanson D, Xu X, Noh SH, Cheong JH, Harismendy O. Prevalence of cancer susceptibility variants in patients with multiple Lynch syndrome related cancers. Sci Rep 2021; 11:14807. [PMID: 34285288 PMCID: PMC8292343 DOI: 10.1038/s41598-021-94292-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022] Open
Abstract
Along with early-onset cancers, multiple primary cancers (MPCs) are likely resulting from increased genetic susceptibility; however, the associated predisposition genes or prevalence of the pathogenic variants genes in MPC patients are often unknown. We screened 71 patients with MPC of the stomach, colorectal, and endometrium, sequencing 65 cancer predisposition genes. A subset of 19 patients with early-onset MPC of stomach and colorectum were further evaluated for variants in cancer related genes using both normal and tumor whole exome sequencing. Among 71 patients with MPCs, variants classified to be pathogenic were observed in 15 (21.1%) patients and affected Lynch Syndrome (LS) genes: MLH1 (n = 10), MSH6 (n = 2), PMS2 (n = 2), and MSH2 (n = 1). All carriers had tumors with high microsatellite instability and 13 of them (86.7%) were early-onset, consistent with LS. In 19 patients with early-onset MPCs, loss of function (LoF) variants in RECQL5 were more prevalent in non-LS MPC than in matched sporadic cancer patients (OR = 31.6, 2.73–1700.6, p = 0.001). Additionally, there were high-confidence LoF variants at FANCG and CASP8 in two patients accompanied by somatic loss of heterozygosity in tumor, respectively. The results suggest that genetic screening should be considered for synchronous cancers and metachronous MPCs of the LS tumor spectrum, particularly in early-onset. Susceptibility variants in non-LS genes for MPC patients may exist, but evidence for their role is more elusive than for LS patients.
Collapse
Affiliation(s)
- Yoon Young Choi
- Department of Surgery, CHA University School of Medicine, Pocheon-si, Korea.,Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu,, Seoul, 120-752, Korea.,Yonsei Biomedical Research Institute, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Jin Shin
- Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Jae Eun Lee
- Yonsei Biomedical Research Institute, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Lisa Madlensky
- Moores Cancer Center and Division of Biomedical Informatics Department of Medicine, University of California San Diego School of Medicine, 3855 Health Sciences Dr, La Jolla, CA, 92037, USA.,Department of Family Medicine and Public Health, University of California San Diego School of Medicine, San Diego, CA, USA
| | - Seung-Tae Lee
- Hereditary Cancer Clinic, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea.,Department of Laboratory Medicine, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Soo Park
- Hereditary Cancer Clinic, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea.,Department of Medicine, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong-Hyeon Jo
- Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea
| | - Daniela Nachmanson
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego School of Medicine, San Diego, USA
| | - Xiaojun Xu
- Moores Cancer Center and Division of Biomedical Informatics Department of Medicine, University of California San Diego School of Medicine, 3855 Health Sciences Dr, La Jolla, CA, 92037, USA
| | - Sung Hoon Noh
- Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu,, Seoul, 120-752, Korea
| | - Jae-Ho Cheong
- Department of Surgery, Yonsei University Health System, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu,, Seoul, 120-752, Korea. .,Yonsei Biomedical Research Institute, Yonsei University Health System, Yonsei University College of Medicine, Seoul, Korea.
| | - Olivier Harismendy
- Moores Cancer Center and Division of Biomedical Informatics Department of Medicine, University of California San Diego School of Medicine, 3855 Health Sciences Dr, La Jolla, CA, 92037, USA. .,Department of Medicine, University of California San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
21
|
Zeng B, Huang P, Du P, Sun X, Huang X, Fang X, Li L. Comprehensive Study of Germline Mutations and Double-Hit Events in Esophageal Squamous Cell Cancer. Front Oncol 2021; 11:637431. [PMID: 33889545 PMCID: PMC8056176 DOI: 10.3389/fonc.2021.637431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/10/2021] [Indexed: 01/12/2023] Open
Abstract
Esophageal squamous cell cancer (ESCC) is the eighth most common cancer around the world. Several reports have focused on somatic mutations and common germline mutations in ESCC. However, the contributions of pathogenic germline alterations in cancer susceptibility genes (CSGs), highly frequently mutated CSGs, and pathogenically mutated CSG-related pathways in ESCC remain unclear. We obtained data on 571 ESCC cases from public databases and East Asian from the 1000 Genomes Project database and the China Metabolic Analytics Project database to characterize pathogenic mutations. We detected 157 mutations in 75 CSGs, accounting for 25.0% (143/571) of ESCC cases. Six genes had more than five mutations: TP53 (n = 15 mutations), GJB2 (n = 8), BRCA2 (n = 6), RECQL4 (n = 6), MUTYH (n = 6), and PMS2 (n = 5). Our results identified significant differences in pathogenic germline mutations of TP53, BRCA2, and RECQL4 between the ESCC and control cohorts. Moreover, we identified 84 double-hit events (16 germline/somatic double-hit events and 68 somatic/somatic double-hit events) occurring in 18 tumor suppressor genes from 83 patients. Patients who had ESCC with germline/somatic double-hit events were diagnosed at younger ages than patients with the somatic/somatic double-hit events, though the correlation was not significant. Fanconi anemia was the most enriched pathway of pathogenically mutated CSGs, and it appeared to be a primary pathway for ESCC predisposition. The results of this study identified the underlying roles that pathogenic germline mutations in CSGs play in ESCC pathogenesis, increased our awareness about the genetic basis of ESCC, and provided suggestions for using highly mutated CSGs and double-hit features in the early discovery, prevention, and genetic counseling of ESCC.
Collapse
Affiliation(s)
- Bing Zeng
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
- BGI-Shenzhen, Shenzhen, China
| | | | - Peina Du
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | | | | | - Xiaodong Fang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Lin Li
- BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
22
|
Mosquera Orgueira A, Cid López M, Peleteiro Raíndo A, Díaz Arias JÁ, Antelo Rodríguez B, Bao Pérez L, Alonso Vence N, Bendaña López Á, Abuin Blanco A, Melero Valentín P, Ferreiro Ferro R, Aliste Santos C, Fraga Rodríguez MF, González Pérez MS, Pérez Encinas MM, Bello López JL. Detection of Rare Germline Variants in the Genomes of Patients with B-Cell Neoplasms. Cancers (Basel) 2021; 13:cancers13061340. [PMID: 33809641 PMCID: PMC8001490 DOI: 10.3390/cancers13061340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The global importance of rare variants in tumorigenesis has been addressed by some pan-cancer analysis, revealing significant enrichments in protein-truncating variants affecting genes such as ATM, BRCA1/2, BRIP1, and MSH6. Germline variants can influence treatment response and contribute to the development of treatment-related second neoplasms, especially in childhood leukemia. We aimed to analyze the genomes of patients with B-cell lymphoproliferative disorders for the discovery of genes enriched in rare pathogenic variants. We discovered a significant enrichment for two genes in germline rare and dysfunctional variants. Additionally, we detected rare and likely pathogenic variants associated with disease prognosis and potential druggability, indicating a relevant role of these events in the variability of cancer phenotypes. Abstract There is growing evidence indicating the implication of germline variation in cancer predisposition and prognostication. Here, we describe an analysis of likely disruptive rare variants across the genomes of 726 patients with B-cell lymphoid neoplasms. We discovered a significant enrichment for two genes in rare dysfunctional variants, both of which participate in the regulation of oxidative stress pathways (CHMP6 and GSTA4). Additionally, we detected 1675 likely disrupting variants in genes associated with cancer, of which 44.75% were novel events and 7.88% were protein-truncating variants. Among these, the most frequently affected genes were ATM, BIRC6, CLTCL1A, and TSC2. Homozygous or germline double-hit variants were detected in 28 cases, and coexisting somatic events were observed in 17 patients, some of which affected key lymphoma drivers such as ATM, KMT2D, and MYC. Finally, we observed that variants in six different genes were independently associated with shorter survival in CLL. Our study results support an important role for rare germline variation in the pathogenesis and prognosis of B-cell lymphoid neoplasms.
Collapse
Affiliation(s)
- Adrián Mosquera Orgueira
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
- Correspondence: ; Tel.: +34-981-950-191
| | - Miguel Cid López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Andrés Peleteiro Raíndo
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - José Ángel Díaz Arias
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Beatriz Antelo Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Laura Bao Pérez
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Natalia Alonso Vence
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Ángeles Bendaña López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Aitor Abuin Blanco
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Paula Melero Valentín
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Roi Ferreiro Ferro
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Carlos Aliste Santos
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - Máximo Francisco Fraga Rodríguez
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
- Department of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Marta Sonia González Pérez
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
| | - Manuel Mateo Pérez Encinas
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Pathology, SERGAS, 15706 Santiago de Compostela, Spain;
| | - José Luis Bello López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.C.L.); (A.P.R.); (J.Á.D.A.); (B.A.R.); (N.A.V.); (Á.B.L.); (M.F.F.R.); (M.S.G.P.); (M.M.P.E.); (J.L.B.L.)
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Department of Hematology, SERGAS, 15706 Santiago de Compostela, Spain; (L.B.P.); (A.A.B.); (P.M.V.); (R.F.F.)
- Department of Medicine, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
23
|
Soares de Lima Y, Arnau-Collell C, Díaz-Gay M, Bonjoch L, Franch-Expósito S, Muñoz J, Moreira L, Ocaña T, Cuatrecasas M, Herrera-Pariente C, Carballal S, Moreno L, Díaz de Bustamante A, Castells A, Bujanda L, Cubiella J, Rodríguez-Alcalde D, Balaguer F, Castellví-Bel S. Germline and Somatic Whole-Exome Sequencing Identifies New Candidate Genes Involved in Familial Predisposition to Serrated Polyposis Syndrome. Cancers (Basel) 2021; 13:929. [PMID: 33672345 PMCID: PMC7927050 DOI: 10.3390/cancers13040929] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
The serrated polyposis syndrome (SPS) is the most common and yet underdiagnosed colorectal polyposis syndrome. It is characterized by multiple and/or large colonic serrated polyps and a higher associated risk for colorectal cancer (CRC). The main objective of this study was to identify new candidate genes involved in the germline predisposition to SPS/CRC. Thirty-nine SPS patients from 16 families (≥2 patients per family) were recruited without alterations in well-known hereditary CRC genes, and germline and somatic whole-exome sequencing were performed. Germline rare variants with plausible pathogenicity, located in genes involved in cancer development, senescence and epigenetic regulation were selected. Somatic mutational profiling and signature analysis was pursued in one sample per family, when possible. After data filtering, ANXA10, ASXL1, CFTR, DOT1L, HIC1, INO80, KLF3, MCM3AP, MCM8, PDLIM2, POLD1, TP53BP1, WNK2 and WRN were highlighted as the more promising candidate genes for SPS germline predisposition with potentially pathogenic variants shared within families. Somatic analysis characterized mutational profiles in advanced serrated polyps/tumors, revealing a high proportion of hypermutated samples, with a prevalence of clock-like mutational signatures in most samples and the presence of DNA mismatch repair-defective signatures in some cases. In conclusion, we identified new candidate genes to be involved in familial SPS. Further functional studies and replication in additional cohorts are required to confirm the selected candidates.
Collapse
Affiliation(s)
- Yasmin Soares de Lima
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Coral Arnau-Collell
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Marcos Díaz-Gay
- Moores Cancer Center, Department of Cellular and Molecular Medicine, Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Laia Bonjoch
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Sebastià Franch-Expósito
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Jenifer Muñoz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Leticia Moreira
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Teresa Ocaña
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Miriam Cuatrecasas
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Pathology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Tumor Bank-Biobank, Hospital Clínic, 08036 Barcelona, Spain;
| | - Cristina Herrera-Pariente
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Sabela Carballal
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Lorena Moreno
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | | | - Antoni Castells
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Luis Bujanda
- Gastroenterology Department, Hospital Donostia-Instituto Biodonostia, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Basque Country University (UPV/EHU), 20014 San Sebastián, Spain;
| | - Joaquín Cubiella
- Gastroenterology Department, Complexo Hospitalario Universitario de Ourense, Instituto de Investigación Sanitaria Galicia Sur, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 32005 Ourense, Spain;
| | | | - Francesc Balaguer
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| | - Sergi Castellví-Bel
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, 08036 Barcelona, Spain; (Y.S.d.L.); (C.A.-C.); (L.B.); (S.F.-E.); (J.M.); (L.M.); (T.O.); (C.H.-P.); (S.C.); (L.M.); (A.C.); (F.B.)
| |
Collapse
|
24
|
Elcheva IA, Spiegelman VS. Targeting RNA-binding proteins in acute and chronic leukemia. Leukemia 2021; 35:360-376. [PMID: 33149266 PMCID: PMC7868134 DOI: 10.1038/s41375-020-01066-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/28/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023]
Abstract
RNA-binding proteins (RBPs) play a crucial role in cellular physiology by regulating RNA processing, translation, and turnover. In neoplasms, RBP support of cancer-relevant expression of alternatively spliced, modified, and stabilized mRNA transcripts is essential to self-renewal, proliferation, and adaptation to stress. In this review, we assess the impact of key families of RBPs in leukemogenesis, review progress in targeting those proteins with small molecules, and discuss how multilevel composition of posttranscriptional regulation of gene expression could be used for potential therapies in acute and chronic leukemia.
Collapse
Affiliation(s)
- Irina A Elcheva
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Vladimir S Spiegelman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
25
|
Milanese JS, Wang E. Germline Genetics in Cancer: The New Frontier. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11667-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
26
|
How wide is the application of genetic big data in biomedicine. Biomed Pharmacother 2020; 133:111074. [PMID: 33378973 DOI: 10.1016/j.biopha.2020.111074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/16/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022] Open
Abstract
In the era of big data, massive genetic data, as a new industry, has quickly swept almost all industries, especially the pharmaceutical industry. As countries around the world start to build their own gene banks, scientists study the data to explore the origins and migration of humans. Moreover, big data encourage the development of cancer therapy and bring good news to cancer patients. Big datum has been involved in the study of many diseases, and it has been found that analyzing diseases at the gene level can lead to more beneficial treatment options than ordinary treatments. This review will introduce the development of extensive data in medical research from the perspective of big data and tumor, neurological and psychiatric diseases, cardiovascular diseases, other applications and the development direction of big data in medicine.
Collapse
|
27
|
Postema FAM, Oosterwijk JC, Hennekam RC. Genetic control of tumor development in malformation syndromes. Am J Med Genet A 2020; 185:324-335. [PMID: 33141500 DOI: 10.1002/ajmg.a.61947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 02/01/2023]
Abstract
One of the questions that arises frequently when caring for an individual with a malformation syndrome, is whether some form of tumor surveillance is indicated. In some syndromes there is a highly variable increased risk to develop tumors, while in others this is not the case. The risks can be hard to predict and difficult to explain to affected individuals and their families, and often also to caregivers. The queries arise especially if syndrome causing mutations are also known to occur in tumors. It needs insight in the mechanisms to understand and explain differences of tumor occurrence, and to offer optimal care to individuals with syndromes. Here we provide a short overview of the major mechanisms of the control for tumor occurrences in malformation syndromes.
Collapse
Affiliation(s)
- Floor A M Postema
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan C Oosterwijk
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Raoul C Hennekam
- Department of Pediatrics, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Zhang S, DuBois W, Zhang K, Simmons JK, Hughitt VK, Gorjifard S, Gaikwad S, Peat TJ, Mock BA. Mouse tumor susceptibility genes identify drug combinations for multiple myeloma. ACTA ACUST UNITED AC 2020; 6. [PMID: 32923678 PMCID: PMC7486007 DOI: 10.20517/2394-4722.2020.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Long-term genetic studies utilizing backcross and congenic strain analyses coupled with positional cloning strategies and functional studies identified Cdkn2a, Mtor, and Mndal as mouse plasmacytoma susceptibility/resistance genes. Tumor incidence data in congenic strains carrying the resistance alleles of Cdkn2a and Mtor led us to hypothesize that drug combinations affecting these pathways are likely to have an additive, if not synergistic effect in inhibiting tumor cell growth. Traditional and novel systems-level genomic approaches were used to assess combination activity, disease specificity, and clinical potential of a drug combination involving rapamycin/everolimus, an Mtor inhibitor, with entinostat, an histone deacetylase inhibitor. The combination synergistically repressed oncogenic MYC and activated the Cdkn2a tumor suppressor. The identification of MYC as a primary upstream regulator led to the identification of small molecule binders of the G-quadruplex structure that forms in the NHEIII region of the MYC promoter. These studies highlight the importance of identifying drug combinations which simultaneously upregulate tumor suppressors and downregulate oncogenes.
Collapse
Affiliation(s)
- Shuling Zhang
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Wendy DuBois
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Ke Zhang
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - John K Simmons
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA.,Personal Genome Diagnostics, Baltimore, MD 21224, USA
| | - V Keith Hughitt
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Sayeh Gorjifard
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA.,University of Washington School of Medicine, Department of Genome Sciences, Seattle, WA 98195, USA
| | - Snehal Gaikwad
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Tyler J Peat
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
29
|
Meggendorfer M, Walter W, Haferlach T. WGS and WTS in leukaemia: A tool for diagnostics? Best Pract Res Clin Haematol 2020; 33:101190. [DOI: 10.1016/j.beha.2020.101190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
|
30
|
Germline genomes have a dominant-heritable contribution to cancer immune evasion and immunotherapy response. QUANTITATIVE BIOLOGY 2020. [DOI: 10.1007/s40484-020-0212-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
31
|
Leonards K, Almosailleakh M, Tauchmann S, Bagger FO, Thirant C, Juge S, Bock T, Méreau H, Bezerra MF, Tzankov A, Ivanek R, Losson R, Peters AHFM, Mercher T, Schwaller J. Nuclear interacting SET domain protein 1 inactivation impairs GATA1-regulated erythroid differentiation and causes erythroleukemia. Nat Commun 2020; 11:2807. [PMID: 32533074 PMCID: PMC7293310 DOI: 10.1038/s41467-020-16179-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 04/17/2020] [Indexed: 12/20/2022] Open
Abstract
The nuclear receptor binding SET domain protein 1 (NSD1) is recurrently mutated in human cancers including acute leukemia. We show that NSD1 knockdown alters erythroid clonogenic growth of human CD34+ hematopoietic cells. Ablation of Nsd1 in the hematopoietic system of mice induces a transplantable erythroleukemia. In vitro differentiation of Nsd1−/− erythroblasts is majorly impaired despite abundant expression of GATA1, the transcriptional master regulator of erythropoiesis, and associated with an impaired activation of GATA1-induced targets. Retroviral expression of wildtype NSD1, but not a catalytically-inactive NSD1N1918Q SET-domain mutant induces terminal maturation of Nsd1−/− erythroblasts. Despite similar GATA1 protein levels, exogenous NSD1 but not NSDN1918Q significantly increases the occupancy of GATA1 at target genes and their expression. Notably, exogenous NSD1 reduces the association of GATA1 with the co-repressor SKI, and knockdown of SKI induces differentiation of Nsd1−/− erythroblasts. Collectively, we identify the NSD1 methyltransferase as a regulator of GATA1-controlled erythroid differentiation and leukemogenesis. Loss of function mutations of NSD1 occur in blood cancers. Here, the authors report that NSD1 loss blocks erythroid differentiation which leads to an erythroleukemia-like disease in mice by impairing GATA1-induced target gene activation.
Collapse
Affiliation(s)
- Katharina Leonards
- University Children's Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Marwa Almosailleakh
- University Children's Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Samantha Tauchmann
- University Children's Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Frederik Otzen Bagger
- University Children's Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland.,Swiss Institute of Bioinfomatics, 4031, Basel, Switzerland.,Genomic Medicine, Righospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Cécile Thirant
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, Gustave Roussy Institute, Université Paris Diderot, Université Paris-Sud, Villejuif, 94800, France
| | - Sabine Juge
- University Children's Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum University of Basel, Basel, Switzerland
| | - Hélène Méreau
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Matheus F Bezerra
- University Children's Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland.,Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, Brazil
| | - Alexandar Tzankov
- Institute for Pathology, University Hospital Basel, 4031, Basel, Switzerland
| | - Robert Ivanek
- Department of Biomedicine, University of Basel, 4031, Basel, Switzerland.,Swiss Institute of Bioinfomatics, 4031, Basel, Switzerland
| | - Régine Losson
- Institute de Génétique et de Biologie Moléculaire et Cellulaire (I.G.B.M.C.), CNRS/INSERM Université de Strasbourg, BP10142, 67404, Illkirch Cedex, France
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research, 4058, Basel, Switzerland.,Faculty of Sciences, University of Basel, 4056, Basel, Switzerland
| | - Thomas Mercher
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, Gustave Roussy Institute, Université Paris Diderot, Université Paris-Sud, Villejuif, 94800, France
| | - Juerg Schwaller
- University Children's Hospital Basel, Basel, Switzerland. .,Department of Biomedicine, University of Basel, 4031, Basel, Switzerland.
| |
Collapse
|
32
|
Oak N, Cherniack AD, Mashl RJ, Hirsch FR, Ding L, Beroukhim R, Gümüş ZH, Plon SE, Huang KL. Ancestry-specific predisposing germline variants in cancer. Genome Med 2020; 12:51. [PMID: 32471518 PMCID: PMC7260738 DOI: 10.1186/s13073-020-00744-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/07/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Distinct prevalence of inherited genetic predisposition may partially explain the difference of cancer risks across ancestries. Ancestry-specific analyses of germline genomes are required to inform cancer genetic risk and prognosis of diverse populations. METHODS We conducted analyses using germline and somatic sequencing data generated by The Cancer Genome Atlas. Collapsing pathogenic and likely pathogenic variants to cancer predisposition genes (CPG), we analyzed the association between CPGs and cancer types within ancestral groups. We also identified the predisposition-associated two-hit events and gene expression effects in tumors. RESULTS Genetic ancestry analysis classified the cohort of 9899 cancer cases into individuals of primarily European (N = 8184, 82.7%), African (N = 966, 9.8%), East Asian (N = 649, 6.6%), South Asian (N = 48, 0.5%), Native/Latin American (N = 41, 0.4%), and admixed (N = 11, 0.1%) ancestries. In the African ancestry, we discovered a potentially novel association of BRCA2 in lung squamous cell carcinoma (OR = 41.4 [95% CI, 6.1-275.6]; FDR = 0.002) previously identified in Europeans, along with a known association of BRCA2 in ovarian serous cystadenocarcinoma (OR = 8.5 [95% CI, 1.5-47.4]; FDR = 0.045). In the East Asian ancestry, we discovered one previously known association of BRIP1 in stomach adenocarcinoma (OR = 12.8 [95% CI, 1.8-90.8]; FDR = 0.038). Rare variant burden analysis further identified 7 suggestive associations in African ancestry individuals previously described in European ancestry, including SDHB in pheochromocytoma and paraganglioma, ATM in prostate adenocarcinoma, VHL in kidney renal clear cell carcinoma, FH in kidney renal papillary cell carcinoma, and PTEN in uterine corpus endometrial carcinoma. Most predisposing variants were found exclusively in one ancestry in the TCGA and gnomAD datasets. Loss of heterozygosity was identified for 7 out of the 15 African ancestry carriers of predisposing variants. Further, tumors from the SDHB or BRCA2 carriers showed simultaneous allelic-specific expression and low gene expression of their respective affected genes, and FH splice-site variant carriers showed mis-splicing of FH. CONCLUSIONS While several CPGs are shared across patients, many pathogenic variants are found to be ancestry-specific and trigger somatic effects. Studies using larger cohorts of diverse ancestries are required to pinpoint ancestry-specific genetic predisposition and inform genetic screening strategies.
Collapse
Affiliation(s)
- Ninad Oak
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrew D Cherniack
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - R Jay Mashl
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63108, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
| | - Fred R Hirsch
- Department of Oncological Sciences, Center for Thoracic Oncology, Tisch Cancer Institute, New York, NY, USA
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, 63108, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, 63108, USA
- Department of Genetics, Washington University in St. Louis, St. Louis, MO, 63108, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, 63108,, USA
| | - Rameen Beroukhim
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sharon E Plon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
33
|
Bioinformatic Analysis Reveals an Immune/Inflammatory-Related Risk Signature for Oral Cavity Squamous Cell Carcinoma. JOURNAL OF ONCOLOGY 2019; 2019:3865279. [PMID: 31911802 PMCID: PMC6930791 DOI: 10.1155/2019/3865279] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/06/2019] [Accepted: 11/05/2019] [Indexed: 12/26/2022]
Abstract
High-throughput gene expression profiling has recently emerged as a promising technique that provides insight into cancer subtype classification and improved prediction of prognoses. Immune/inflammatory-related mRNAs may potentially enrich genes to allow researchers to better illustrate cancer microenvironments. Oral cavity squamous cell carcinoma (OC-SCC) exhibits high morbidity and poor prognosis compared to that of other types of head and neck squamous cell carcinoma (HNSCC), and these differences may be partially due to differences within the tumor microenvironments. Based on this, we designed an immune-related signature to improve the prognostic prediction of OC-SCC. A cohort of 314 OC-SCC samples possessing whole genome expression data that were sourced from The Cancer Genome Atlas (TCGA) database was included for discovery. The GSE41613 database was used for validation. A risk score was established using immune/inflammatory signatures acquired from the training dataset. Principal components analysis, GO analysis, and gene set enrichment analysis were used to explore the bioinformatic implications. When grouped by the dichotomized risk score based on the signature, this classifier could successfully discriminate patients with distinct prognoses within the training and validation cohorts (P < 0.05 in both cohorts) and within different clinicopathological subgroups. Similar somatic mutation patterns were observed between high and low risk score groups, and different copy number variation patterns were also identified. Further bioinformatic analyses suggested that the lower risk score group was significantly correlated with immune/inflammatory-related biological processes, while the higher risk score group was highly associated with cell cycle-related processes. The analysis indicated that the risk score was a robust predictor of patient survival, and its functional annotation was well established. Therefore, this bioinformatic-based immune-related signature suggested that the microenvironment of OC-SCC could distinguish among patients with different underlying biological processes and clinical outcomes, and the use of this signature may shed light on future OC-SCC classification and therapeutic design.
Collapse
|
34
|
Mandelker D, Ceyhan-Birsoy O. Evolving Significance of Tumor-Normal Sequencing in Cancer Care. Trends Cancer 2019; 6:31-39. [PMID: 31952779 DOI: 10.1016/j.trecan.2019.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/14/2019] [Accepted: 11/17/2019] [Indexed: 12/15/2022]
Abstract
Molecular tests assist at various stages of cancer patient management, including providing diagnosis, predicting prognosis, identifying therapeutic targets, and determining hereditary cancer risk. The current testing paradigm involves germline testing in a subset of patients determined to be at high risk for having a hereditary cancer syndrome, and tumor-only sequencing for treatment decisions in advanced cancer patients. A major limitation of tumor-only sequencing is its inability to distinguish germline versus somatic mutations. Tumor-normal sequencing has emerged as a comprehensive analysis for both hereditary cancer predisposition and somatic profiling. Here, we review recent studies involving tumor-normal sequencing, discuss its benefits in clinical care, challenges for its implementation, and novel insights it has provided regarding tumor biology and germline contribution to cancer.
Collapse
Affiliation(s)
- Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Ozge Ceyhan-Birsoy
- Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
35
|
Cheng YI, Gan YC, Liu D, Davies MPA, Li WM, Field JK. Potential genetic modifiers for somatic EGFR mutation in lung cancer: a meta-analysis and literature review. BMC Cancer 2019; 19:1068. [PMID: 31703574 PMCID: PMC6842246 DOI: 10.1186/s12885-019-6317-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/31/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Accumulating evidence indicates inherited risk in the aetiology of lung cancer, although smoking exposure is the major attributing factor. Family history is a simple substitute for inherited susceptibility. Previous studies have shown some possible yet conflicting links between family history of cancer and EGFR mutation in lung cancer. As EGFR-mutated lung cancer favours female, never-smoker, adenocarcinoma and Asians, it may be argued that there may be some underlying genetic modifiers responsible for the pathogenesis of EGFR mutation. METHODS We searched four databases for all original articles on family history of malignancy and EGFR mutation status in lung cancer published up to July 2018. We performed a meta-analysis by using a random-effects model and odds ratio estimates. Heterogeneity and sensitivity were also investigated. Then we conducted a second literature research to curate case reports of familial lung cancers who studied both germline cancer predisposing genes and their somatic EGFR mutation status; and explored the possible links between cancer predisposing genes and EGFR mutation. RESULTS Eleven studies have been included in the meta-analysis. There is a significantly higher likelihood of EGFR mutation in lung cancer patients with family history of cancer than their counterparts without family history, preferentially in Asians (OR = 1.35[1.06-1.71], P = 0.01), those diagnosed with adenocarcinomas ((OR = 1.47[1.14-1.89], P = 0.003) and those with lung cancer-affected relatives (first and second-degree: OR = 1.53[1.18-1.99], P = 0.001; first-degree: OR = 1.76[1.36-2.28, P < 0.0001]). Familial lung cancers more likely have concurrent EGFR mutations along with mutations in their germline cancer predisposition genes including EGFR T790 M, BRCA2 and TP53. Certain mechanisms may contribute to the combination preferences between inherited mutations and somatic ones. CONCLUSIONS Potential genetic modifiers may contribute to somatic EGFR mutation in lung cancer, although current data is limited. Further studies on this topic are needed, which may help to unveil lung carcinogenesis pathways. However, caution is warranted in data interpretation due to limited cases available for the current study.
Collapse
Affiliation(s)
- Yue I Cheng
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
- Lung Cancer Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Yun Cui Gan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Michael P A Davies
- Lung Cancer Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Wei Min Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - John K Field
- Lung Cancer Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| |
Collapse
|
36
|
Abdi E, Latifi‐Navid S, Zahri S, Yazdanbod A, Pourfarzi F. Risk factors predisposing to cardia gastric adenocarcinoma: Insights and new perspectives. Cancer Med 2019; 8:6114-6126. [PMID: 31448582 PMCID: PMC6792520 DOI: 10.1002/cam4.2497] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/17/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Recent decades have seen an alarming increase in the incidence of cardia gastric adenocarcinoma (CGA) while noncardia gastric adenocarcinoma (NCGA) has decreased. In 2012, 260 000 CGA cases (age-standardised rate (ASR); 3.3/100 000) and 691 000 NCGA cases (ASR; 8.8/100 000) were reported worldwide. Compared with women, men had greater rates for both the subsites, especially for CGA. Recently, four molecular subtypes of GC have been proposed by the Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG); however, these classifications do not take into account predisposing germline variants and their possible interaction with somatic alterations in carcinogenesis. The etiology of adenocarcinoma of the cardia and the gastroesophageal junction (GEJ) is not known. It is thought that CGA is distinct from adenocarcinomas located in the esophagus or distal stomach, both epidemiologically and biologically. Moreover, CGA is often identified in the advanced stage having a poor prognosis. Therefore, understanding the risk and the role of predisposing factors in etiology of CGA can inform clinical practice and counseling for risk reduction. In this paper, we showed that GC family history, lifestyle, demographics, gastroesophageal reflux disease, Helicobacter pylori infection, and multiple genetic and epigenetic risk factors as well as several predisposing conditions may underlie susceptibility to CGA. However, several genome-wide association studies (GWASs) should be conducted to identify novel high-penetrance genes and pathways as well as causal germline variants predisposing to CGA. They must include different ethnic groups, especially from high-incidence countries for CGA, because some risk loci are ancestry-specific. In parallel, statistical methods can be developed to identify cancer predisposition genes (CPGs) from tumor sequencing data. It is also necessary to find novel long noncoding RNAs related to the risk of CGA. Taken altogether, new cancer risk prediction models, including all genetic and nongenetic factors influencing risk, should be developed to facilitate risk assessment, disease prevention, and early diagnosis and intervention of CGA in the future.
Collapse
Affiliation(s)
- Esmat Abdi
- Department of BiologyFaculty of SciencesUniversity of Mohaghegh ArdabiliArdabilIran
| | - Saeid Latifi‐Navid
- Department of BiologyFaculty of SciencesUniversity of Mohaghegh ArdabiliArdabilIran
| | - Saber Zahri
- Department of BiologyFaculty of SciencesUniversity of Mohaghegh ArdabiliArdabilIran
| | - Abbas Yazdanbod
- Digestive Diseases Research CenterArdabil University of Medical SciencesArdabilIran
| | - Farhad Pourfarzi
- Digestive Diseases Research CenterArdabil University of Medical SciencesArdabilIran
| |
Collapse
|
37
|
Rasnic R, Brandes N, Zuk O, Linial M. Substantial batch effects in TCGA exome sequences undermine pan-cancer analysis of germline variants. BMC Cancer 2019; 19:783. [PMID: 31391007 PMCID: PMC6686424 DOI: 10.1186/s12885-019-5994-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 07/30/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In recent years, research on cancer predisposition germline variants has emerged as a prominent field. The identity of somatic mutations is based on a reliable mapping of the patient germline variants. In addition, the statistics of germline variants frequencies in healthy individuals and cancer patients is the basis for seeking candidates for cancer predisposition genes. The Cancer Genome Atlas (TCGA) is one of the main sources of such data, providing a diverse collection of molecular data including deep sequencing for more than 30 types of cancer from > 10,000 patients. METHODS Our hypothesis in this study is that whole exome sequences from blood samples of cancer patients are not expected to show systematic differences among cancer types. To test this hypothesis, we analyzed common and rare germline variants across six cancer types, covering 2241 samples from TCGA. In our analysis we accounted for inherent variables in the data including the different variant calling protocols, sequencing platforms, and ethnicity. RESULTS We report on substantial batch effects in germline variants associated with cancer types. We attribute the effect to the specific sequencing centers that produced the data. Specifically, we measured 30% variability in the number of reported germline variants per sample across sequencing centers. The batch effect is further expressed in nucleotide composition and variant frequencies. Importantly, the batch effect causes substantial differences in germline variant distribution patterns across numerous genes, including prominent cancer predisposition genes such as BRCA1, RET, MAX, and KRAS. For most of known cancer predisposition genes, we found a distinct batch-dependent difference in germline variants. CONCLUSION TCGA germline data is exposed to strong batch effects with substantial variabilities among TCGA sequencing centers. We claim that those batch effects are consequential for numerous TCGA pan-cancer studies. In particular, these effects may compromise the reliability and the potency to detect new cancer predisposition genes. Furthermore, interpretation of pan-cancer analyses should be revisited in view of the source of the genomic data after accounting for the reported batch effects.
Collapse
Affiliation(s)
- Roni Rasnic
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Nadav Brandes
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Or Zuk
- Department of Statistics, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Linial
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
38
|
Miller JE, Metpally RP, Person TN, Krishnamurthy S, Dasari VR, Shivakumar M, Lavage DR, Cook AM, Carey DJ, Ritchie MD, Kim D, Gogoi R. Systematic characterization of germline variants from the DiscovEHR study endometrial carcinoma population. BMC Med Genomics 2019; 12:59. [PMID: 31053132 PMCID: PMC6499978 DOI: 10.1186/s12920-019-0504-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/15/2019] [Indexed: 02/02/2023] Open
Abstract
Background Endometrial cancer (EMCA) is the fifth most common cancer among women in the world. Identification of potentially pathogenic germline variants from individuals with EMCA will help characterize genetic features that underlie the disease and potentially predispose individuals to its pathogenesis. Methods The Geisinger Health System’s (GHS) DiscovEHR cohort includes exome sequencing on over 50,000 consenting patients, 297 of whom have evidence of an EMCA diagnosis in their electronic health record. Here, rare variants were annotated as potentially pathogenic. Results Eight genes were identified as having increased burden in the EMCA cohort relative to the non-cancer control cohort. None of the eight genes had an increased burden in the other hormone related cancer cohort from GHS, suggesting they can help characterize the underlying genetic variation that gives rise to EMCA. Comparing GHS to the cancer genome atlas (TCGA) EMCA germline data illustrated 34 genes with potentially pathogenic variation and eight unique potentially pathogenic variants that were present in both studies. Thus, similar germline variation among genes can be observed in unique EMCA cohorts and could help prioritize genes to investigate for future work. Conclusion In summary, this systematic characterization of potentially pathogenic germline variants describes the genetic underpinnings of EMCA through the use of data from a single hospital system. Electronic supplementary material The online version of this article (10.1186/s12920-019-0504-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jason E Miller
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Raghu P Metpally
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | - Thomas N Person
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | | | | | - Manu Shivakumar
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | - Daniel R Lavage
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA
| | - Adam M Cook
- Weis Center for Research, Geisinger Medical Center, Danville, PA, 17822, USA
| | - David J Carey
- Weis Center for Research, Geisinger Medical Center, Danville, PA, 17822, USA
| | - Marylyn D Ritchie
- Department of Genetics, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dokyoon Kim
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA, 17822, USA.,Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, USA
| | - Radhika Gogoi
- Weis Center for Research, Geisinger Medical Center, Danville, PA, 17822, USA.
| | | |
Collapse
|
39
|
Plichta JK, Sebastian ML, Smith LA, Menendez CS, Johnson AT, Bays SM, Euhus DM, Clifford EJ, Jalali M, Kurtzman SH, Taylor WA, Hughes KS. Germline Genetic Testing: What the Breast Surgeon Needs to Know. Ann Surg Oncol 2019; 26:2184-2190. [PMID: 30941656 DOI: 10.1245/s10434-019-07341-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Indexed: 01/21/2023]
Abstract
PURPOSE The American Society of Breast Surgeons (ASBrS) sought to provide educational guidelines for breast surgeons on how to incorporate genetic information and genomics into their practice. METHODS A comprehensive nonsystematic review was performed of selected peer-reviewed literature. The Genetics Working Group of the ASBrS convened to develop guideline recommendations. RESULTS Clinical and educational guidelines were prepared to outline the essential knowledge for breast surgeons to perform germline genetic testing and to incorporate the findings into their practice, which have been approved by the ASBrS Board of Directors. RECOMMENDATIONS Thousands of women in the USA would potentially benefit from genetic testing for BRCA1, BRCA2, and other breast cancer genes that markedly increase their risk of developing breast cancer. As genetic testing is now becoming more widely available, women should be made aware of these tests and consider testing. Breast surgeons are well positioned to help facilitate this process. The areas where surgeons need to be knowledgeable include: (1) identification of patients for initial breast cancer-related genetic testing, (2) identification of patients who tested negative in the past but now need updated testing, (3) initial cancer genetic testing, (4) retesting of patients who need their genetic testing updated, (5) cancer genetic test interpretation, posttest counseling and management, (6) management of variants of uncertain significance, (7) cascade genetic testing, (8) interpretation of genetic tests other than clinical cancer panels and the counseling and management required, and (9) interpretation of somatic genetic tests and the counseling and management required.
Collapse
Affiliation(s)
| | - Molly L Sebastian
- Reinsch Pierce Family Center for Breast Health, Virginia Hospital Center, Arlington, VA, USA
| | | | | | - Anita T Johnson
- Cancer Treatment Centers of America, Atlanta, GA, USA.,Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - David M Euhus
- Department of Surgery, Johns Hopkins University Hospital, Baltimore, MD, USA
| | | | - Mena Jalali
- American Society of Breast Surgeons, Dallas, TX, USA
| | | | | | - Kevin S Hughes
- Department of Surgery, Division of Surgical Oncology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
40
|
Díaz-Gay M, Franch-Expósito S, Arnau-Collell C, Park S, Supek F, Muñoz J, Bonjoch L, Gratacós-Mulleras A, Sánchez-Rojas PA, Esteban-Jurado C, Ocaña T, Cuatrecasas M, Vila-Casadesús M, Lozano JJ, Parra G, Laurie S, Beltran S, EPICOLON Consortium, Castells A, Bujanda L, Cubiella J, Balaguer F, Castellví-Bel S. Integrated Analysis of Germline and Tumor DNA Identifies New Candidate Genes Involved in Familial Colorectal Cancer. Cancers (Basel) 2019; 11:362. [PMID: 30871259 PMCID: PMC6468873 DOI: 10.3390/cancers11030362] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/29/2022] Open
Abstract
Colorectal cancer (CRC) shows aggregation in some families but no alterations in the known hereditary CRC genes. We aimed to identify new candidate genes which are potentially involved in germline predisposition to familial CRC. An integrated analysis of germline and tumor whole-exome sequencing data was performed in 18 unrelated CRC families. Deleterious single nucleotide variants (SNV), short insertions and deletions (indels), copy number variants (CNVs) and loss of heterozygosity (LOH) were assessed as candidates for first germline or second somatic hits. Candidate tumor suppressor genes were selected when alterations were detected in both germline and somatic DNA, fulfilling Knudson's two-hit hypothesis. Somatic mutational profiling and signature analysis were also performed. A series of germline-somatic variant pairs were detected. In all cases, the first hit was presented as a rare SNV/indel, whereas the second hit was either a different SNV (3 genes) or LOH affecting the same gene (141 genes). BRCA2, BLM, ERCC2, RECQL, REV3L and RIF1 were among the most promising candidate genes for germline CRC predisposition. The identification of new candidate genes involved in familial CRC could be achieved by our integrated analysis. Further functional studies and replication in additional cohorts are required to confirm the selected candidates.
Collapse
Affiliation(s)
- Marcos Díaz-Gay
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Sebastià Franch-Expósito
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Coral Arnau-Collell
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Solip Park
- Systems Biology Program, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain;
| | - Fran Supek
- Institut de Recerca Biomedica (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain;
| | - Jenifer Muñoz
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Laia Bonjoch
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Anna Gratacós-Mulleras
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Paula A. Sánchez-Rojas
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Clara Esteban-Jurado
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Teresa Ocaña
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | | | - Maria Vila-Casadesús
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain; (M.V.-C.); (J.J.L.)
| | - Juan José Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain; (M.V.-C.); (J.J.L.)
| | - Genis Parra
- Centre Nacional d’Anàlisi Genòmica-Centre de Regulació Genòmica (CNAG-CRG), Parc Científic de Barcelona, 08028 Barcelona, Spain; (G.P.); (S.L.); (S.B.)
| | - Steve Laurie
- Centre Nacional d’Anàlisi Genòmica-Centre de Regulació Genòmica (CNAG-CRG), Parc Científic de Barcelona, 08028 Barcelona, Spain; (G.P.); (S.L.); (S.B.)
| | - Sergi Beltran
- Centre Nacional d’Anàlisi Genòmica-Centre de Regulació Genòmica (CNAG-CRG), Parc Científic de Barcelona, 08028 Barcelona, Spain; (G.P.); (S.L.); (S.B.)
| | - EPICOLON Consortium
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
- Gastroenterology Department, Hospital Donostia-Instituto Biodonostia, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Basque Country University (UPV/EHU), 20014 San Sebastián, Spain;
- Gastroenterology Department, Complexo Hospitalario Universitario de Ourense, Instituto de Investigación Sanitaria Galicia Sur, 32005 Ourense, Spain;
| | - Antoni Castells
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Luis Bujanda
- Gastroenterology Department, Hospital Donostia-Instituto Biodonostia, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Basque Country University (UPV/EHU), 20014 San Sebastián, Spain;
| | - Joaquín Cubiella
- Gastroenterology Department, Complexo Hospitalario Universitario de Ourense, Instituto de Investigación Sanitaria Galicia Sur, 32005 Ourense, Spain;
| | - Francesc Balaguer
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| | - Sergi Castellví-Bel
- Gastroenterology Department, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Hospital Clínic, 08036 Barcelona, Spain; (M.D.-G.); (S.F.-E.); (C.A.-C.); (J.M.); (L.B.); (A.G.-M.); (P.A.S.-R.); (C.E.-J.); (T.O.); (A.C.); (F.B.)
| |
Collapse
|
41
|
Phatak A, Athar M, Crowell JA, Leffel D, Herbert BS, Bale AE, Kopelovich L. Global gene expression of histologically normal primary skin cells from BCNS subjects reveals "single-hit" effects that are influenced by rapamycin. Oncotarget 2019; 10:1360-1387. [PMID: 30858923 PMCID: PMC6402716 DOI: 10.18632/oncotarget.26640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 01/11/2019] [Indexed: 02/05/2023] Open
Abstract
Studies of dominantly heritable cancers enabled insights about tumor progression. BCNS is a dominantly inherited disorder that is characterized by developmental abnormalities and postnatal neoplasms, principally BCCs. We performed an exploratory gene expression profiling of primary cell cultures derived from clinically unaffected skin biopsies of BCNS gene-carriers (PTCH1+/-) and normal individuals. PCA and HC of untreated keratinocytes or fibroblasts failed to clearly distinguish BCNS samples from controls. These results are presumably due to the common suppression of canonical HH signaling in vitro. We then used a relaxed threshold (p-value <0.05, no FDR cut-off; FC 1.3) that identified a total of 585 and 857 genes differentially expressed in BCNS keratinocytes and fibroblasts samples, respectively. A GSEA identified pancreatic β cell hallmark and mTOR signaling genes in BCNS keratinocytes, whereas analyses of BCNS fibroblasts identified gene signatures regulating pluripotency of stem cells, including WNT pathway. Significantly, rapamycin treatment (FDR<0.05), affected a total of 1411 and 4959 genes in BCNS keratinocytes and BCNS fibroblasts, respectively. In contrast, rapamycin treatment affected a total of 3214 and 4797 genes in normal keratinocytes and normal fibroblasts, respectively. The differential response of BCNS cells to rapamycin involved 599 and 1463 unique probe sets in keratinocytes and fibroblasts, respectively. An IPA of these genes in the presence of rapamycin pointed to hepatic fibrosis/stellate cell activation, and HIPPO signaling in BCNS keratinocytes, whereas mitochondrial dysfunction and AGRN expression were uniquely enriched in BCNS fibroblasts. The gene expression changes seen here are likely involved in the etiology of BCCs and they may represent biomarkers/targets for early intervention.
Collapse
Affiliation(s)
- Amruta Phatak
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - David Leffel
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - Brittney-Shea Herbert
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Allen E Bale
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Levy Kopelovich
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
42
|
Walsh MF, Ritter DI, Kesserwan C, Sonkin D, Chakravarty D, Chao E, Ghosh R, Kemel Y, Wu G, Lee K, Kulkarni S, Hedges D, Mandelker D, Ceyhan-Birsoy O, Luo M, Drazer M, Zhang L, Offit K, Plon SE. Integrating somatic variant data and biomarkers for germline variant classification in cancer predisposition genes. Hum Mutat 2018; 39:1542-1552. [PMID: 30311369 PMCID: PMC6310222 DOI: 10.1002/humu.23640] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022]
Abstract
In its landmark paper about Standards and Guidelines for the Interpretation of Sequence Variants, the American College of Medical Genetics and Genomics (ACMG), and Association for Molecular Pathology (AMP) did not address how to use tumor data when assessing the pathogenicity of germline variants. The Clinical Genome Resource (ClinGen) established a multidisciplinary working group, the Germline/Somatic Variant Subcommittee (GSVS) with this focus. The GSVS implemented a survey to determine current practices of integrating somatic data when classifying germline variants in cancer predisposition genes. The GSVS then reviewed and analyzed available resources of relevant somatic data, and performed integrative germline variant curation exercises. The committee determined that somatic hotspots could be systematically integrated into moderate evidence of pathogenicity (PM1). Tumor RNA sequencing data showing altered splicing may be considered as strong evidence in support of germline pathogenicity (PVS1) and tumor phenotypic features such as mutational signatures be considered supporting evidence of pathogenicity (PP4). However, at present, somatic data such as focal loss of heterozygosity and mutations occurring on the alternative allele are not recommended to be systematically integrated, instead, incorporation of this type of data should take place under the advisement of multidisciplinary cancer center tumor-normal sequencing boards.
Collapse
Affiliation(s)
- Michael F Walsh
- Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | | | | | | | | | | | | | - Yelena Kemel
- Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Gang Wu
- St. Jude Children's Hospital, Memphis, Tennessee, USA
| | - Kristy Lee
- University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Dale Hedges
- St. Jude Children's Hospital, Memphis, Tennessee, USA
| | - Diana Mandelker
- Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | | | - Minjie Luo
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Liying Zhang
- Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | | |
Collapse
|