151
|
Abstract
PALB2 (Partner and Localizer of BRCA2) was first identified as a BRCA2-interacting protein. Subsequently, PALB2 has been recognized as a cog in the cellular machinery for DNA repair by homologous recombination (HR). PALB2 also mediates S and G2 DNA damage checkpoints, and has an apparent function in protecting transcriptionally active genes from genotoxic stress. PALB2 also interacts with, is localized by, and functions downstream of BRCA1. Further, PALB2 interacts with other essential effectors of HR, including RAD51 and RAD51C, as well as BRCA2. Consistent with its function in HR and its interaction with key HR proteins, PALB2-deficient cells are hypersensitive to ionizing radiation and DNA interstrand crosslinking agents such as mitomycin C and cisplatin. Mechanistically, PALB2 is required for HR by mediating the recruitment of BRCA2 and the RAD51 recombinase to sites of DNA damage. Similar to bi-allelic loss-of-function mutations of BRCA1, BRCA2, RAD51 and RAD51C, bi-allelic mutations in PALB2 cause Fanconi anemia (FA), a rare childhood disorder which is associated with progressive bone marrow failure, congenital anomalies, and a predisposition to leukemia and solid tumors. Due to their close functional relationship, bi-allelic mutations of PALB2 and BRCA2 cause particularly severe forms of FA, called FANCN and FANCD1, both characterized by severe congenital abnormalities and very early onset of various cancers. This includes acute leukemias, Wilms tumor, medulloblastoma and neuroblastomas. Also, heterozygous germ-line mutations of PALB2, like mutations in several other essential HR genes listed above, yield an increased susceptibility to breast and pancreatic cancer.
Collapse
Affiliation(s)
- Helmut Hanenberg
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen Germany
| | - Paul R Andreassen
- Division of Experimental Hematology & Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati OH, USA
| |
Collapse
|
152
|
The importance of a well-structured pancreatic screening program for familial and hereditary pancreatic cancer. Fam Cancer 2018; 17:1-3. [PMID: 29204967 DOI: 10.1007/s10689-017-0066-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
153
|
Li D, Wang P, Yu Y, Huang B, Zhang X, Xu C, Zhao X, Yin Z, He Z, Jin M, Liu C. Tumor-preventing activity of aspirin in multiple cancers based on bioinformatic analyses. PeerJ 2018; 6:e5667. [PMID: 30280037 PMCID: PMC6163034 DOI: 10.7717/peerj.5667] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background Acetylsalicylic acid was renamed aspirin in 1899, and it has been widely used for its multiple biological actions. Because of the diversity of the cellular processes and diseases that aspirin reportedly affects and benefits, uncertainty remains regarding its mechanism in different biological systems. Methods The Drugbank and STITCH databases were used to find direct protein targets (DPTs) of aspirin. The Mentha database was used to analyze protein-protein interactions (PPIs) to find DPT-associated genes. DAVID was used for the GO and KEGG enrichment analyses. The cBio Cancer Genomics Portal database was used to mine genetic alterations and networks of aspirin-associated genes in cancer. Results Eighteen direct protein targets (DPT) and 961 DPT-associated genes were identified for aspirin. This enrichment analysis resulted in eight identified KEGG pathways that were associated with cancers. Analysis using the cBio portal indicated that aspirin might have effects on multiple tumor suppressors, such as TP53, PTEN, and RB1 and that TP53 might play a central role in aspirin-associated genes. Discussion The results not only suggest that aspirin might have anti-tumor actions against multiple cancers but could also provide new directions for further research on aspirin using a bioinformatics analysis approach.
Collapse
Affiliation(s)
- Diangeng Li
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| | - Peng Wang
- Chinese PLA General Hospital, Nanlou Medical Oncology Department, Beijing, China
| | - Yi Yu
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| | - Bing Huang
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| | - Xuelin Zhang
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| | - Chou Xu
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| | - Xian Zhao
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| | - Zhiwei Yin
- Hebei Medical University, School of Chinese Integrative Medicine, Shijiazhuang, China
| | - Zheng He
- Chinese PLA General Hospital, Department of Clinical Laboratory, Beijing, China
| | - Meiling Jin
- Beijing Chao-yang Hospital, Department of Nephrology, Beijing, China
| | - Changting Liu
- Chinese PLA General Hospital, Nanlou Respiratory Diseases Department, Beijing, China
| |
Collapse
|
154
|
Regneri J, Klotz B, Wilde B, Kottler VA, Hausmann M, Kneitz S, Regensburger M, Maurus K, Götz R, Lu Y, Walter RB, Herpin A, Schartl M. Analysis of the putative tumor suppressor gene cdkn2ab in pigment cells and melanoma of Xiphophorus and medaka. Pigment Cell Melanoma Res 2018; 32:248-258. [PMID: 30117276 DOI: 10.1111/pcmr.12729] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 01/05/2023]
Abstract
In humans, the CDKN2A locus encodes two transcripts, INK4A and ARF. Inactivation of either one by mutations or epigenetic changes is a frequent signature of malignant melanoma and one of the most relevant entry points for melanomagenesis. To analyze whether cdkn2ab, the fish ortholog of CDKN2A, has a similar function as its human counterpart, we studied its action in fish models for human melanoma. Overexpression of cdkn2ab in a Xiphophorus melanoma cell line led to decreased proliferation and induction of a senescence-like phenotype, indicating a melanoma-suppressive function analogous to mammals. Coexpression of Xiphophorus cdkn2ab in medaka transgenic for the mitfa:xmrk melanoma-inducing gene resulted in full suppression of melanoma development, whereas CRISPR/Cas9 knockout of cdkn2ab resulted in strongly enhanced tumor growth. In summary, this provides the first functional evidence that cdkn2ab acts as a potent tumor suppressor gene in fish melanoma models.
Collapse
Affiliation(s)
- Janine Regneri
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany
| | - Barbara Klotz
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany
| | - Brigitta Wilde
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany
| | - Verena A Kottler
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany
| | - Michael Hausmann
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany
| | - Susanne Kneitz
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany
| | | | - Katja Maurus
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Ralph Götz
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Yuan Lu
- Department of Chemistry & Biochemistry, Molecular Biosciences Research Group, Texas State University, San Marcos, Texas
| | - Ronald B Walter
- Department of Chemistry & Biochemistry, Molecular Biosciences Research Group, Texas State University, San Marcos, Texas
| | - Amaury Herpin
- INRA, Fish Physiology and Genomics Institute (INRA-LPGP), Sexual Differentiation and Oogenesis Group (SDOG), Campus de Beaulieu, Rennes Cedex, France
| | - Manfred Schartl
- Physiological Chemistry, University of Würzburg, Biozentrum, Würzburg, Germany.,Comprehensive Cancer Center, University Clinic Würzburg, Würzburg, Germany.,Hagler Institute for Advanced Study and Department of Biology, Texas A&M University, College Station, Texas
| |
Collapse
|
155
|
Canto MI, Almario JA, Schulick RD, Yeo CJ, Klein A, Blackford A, Shin EJ, Sanyal A, Yenokyan G, Lennon AM, Kamel IR, Fishman EK, Wolfgang C, Weiss M, Hruban RH, Goggins M. Risk of Neoplastic Progression in Individuals at High Risk for Pancreatic Cancer Undergoing Long-term Surveillance. Gastroenterology 2018; 155:740-751.e2. [PMID: 29803839 PMCID: PMC6120797 DOI: 10.1053/j.gastro.2018.05.035] [Citation(s) in RCA: 276] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 04/09/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Screening of individuals who have a high risk of pancreatic ductal adenocarcinoma (PDAC), because of genetic factors, frequently leads to identification of pancreatic lesions. We investigated the incidence of PDAC and risk factors for neoplastic progression in individuals at high risk for PDAC enrolled in a long-term screening study. METHODS We analyzed data from 354 individuals at high risk for PDAC (based on genetic factors of family history), enrolled in Cancer of the Pancreas Screening cohort studies at tertiary care academic centers from 1998 through 2014 (median follow-up time, 5.6 years). All subjects were evaluated at study entry (baseline) by endoscopic ultrasonography and underwent surveillance with endoscopic ultrasonography, magnetic resonance imaging, and/or computed tomography. The primary endpoint was the cumulative incidence of PDAC, pancreatic intraepithelial neoplasia grade 3, or intraductal papillary mucinous neoplasm with high-grade dysplasia (HGD) after baseline. We performed multivariate Cox regression and Kaplan-Meier analyses. RESULTS During the follow-up period, pancreatic lesions with worrisome features (solid mass, multiple cysts, cyst size > 3 cm, thickened/enhancing walls, mural nodule, dilated main pancreatic duct > 5 mm, or abrupt change in duct caliber) or rapid cyst growth (>4 mm/year) were detected in 68 patients (19%). Overall, 24 of 354 patients (7%) had neoplastic progression (14 PDACs and 10 HGDs) over a 16-year period; the rate of progression was 1.6%/year, and 93% had detectable lesions with worrisome features before diagnosis of the PDAC or HGD. Nine of the 10 PDACs detected during routine surveillance were resectable; a significantly higher proportion of patients with resectable PDACs survived 3 years (85%) compared with the 4 subjects with symptomatic, unresectable PDACs (25%), which developed outside surveillance (log rank P < .0001). Neoplastic progression occurred at a median age of 67 years; the median time from baseline screening until PDAC diagnosis was 4.8 years (interquartile range, 1.6-6.9 years). CONCLUSIONS In a long-term (16-year) follow-up study of individuals at high-risk for PDAC, we found most PDACs detected during surveillance (9/10) to be resectable, and 85% of these patients survived for 3 years. We identified radiologic features associated with neoplastic progression.
Collapse
Affiliation(s)
- Marcia Irene Canto
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| | - Jose Alejandro Almario
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions,Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | | | | | - Alison Klein
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Amanda Blackford
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Eun Ji Shin
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Abanti Sanyal
- The Johns Hopkins Biostatistics Center, Johns Hopkins Bloomberg School of Public Health
| | - Gayane Yenokyan
- The Johns Hopkins Biostatistics Center, Johns Hopkins Bloomberg School of Public Health
| | - Anne Marie Lennon
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Ihab R. Kamel
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Elliot K. Fishman
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Christopher Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Matthew Weiss
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Michael Goggins
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions,Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| |
Collapse
|
156
|
Zhan W, Shelton CA, Greer PJ, Brand RE, Whitcomb DC. Germline Variants and Risk for Pancreatic Cancer: A Systematic Review and Emerging Concepts. Pancreas 2018; 47:924-936. [PMID: 30113427 PMCID: PMC6097243 DOI: 10.1097/mpa.0000000000001136] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer requires many genetic mutations. Combinations of underlying germline variants and environmental factors may increase the risk of cancer and accelerate the oncogenic process. We systematically reviewed, annotated, and classified previously reported pancreatic cancer-associated germline variants in established risk genes. Variants were scored using multiple criteria and binned by evidence for pathogenicity, then annotated with published functional studies and associated biological systems/pathways. Twenty-two previously identified pancreatic cancer risk genes and 337 germline variants were identified from 97 informative studies that met our inclusion criteria. Fifteen of these genes contained 66 variants predicted to be pathogenic (APC, ATM, BRCA1, BRCA2, CDKN2A, CFTR, CHEK2, MLH1, MSH2, NBN, PALB2, PALLD, PRSS1, SPINK1, TP53). Pancreatic cancer risk genes were organized into key biological mechanisms that promote pancreatic oncogenesis within an oncogenic model. Development of precision medicine approaches requires updated variant information within the framework of an oncogenic progression model. Complex risk modeling may improve interpretation of early biomarkers and guide pathway-specific treatment for pancreatic cancer in the future. Precision medicine is within reach.
Collapse
Affiliation(s)
- Wei Zhan
- School of Medicine, Tsinghua University, Beijing, China
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Celeste A. Shelton
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Phil J. Greer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Randall E. Brand
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - David C. Whitcomb
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, and University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
157
|
Sehdev A, Gbolahan O, Hancock BA, Stanley M, Shahda S, Wan J, Wu HH, Radovich M, O’Neil BH. Germline and Somatic DNA Damage Repair Gene Mutations and Overall Survival in Metastatic Pancreatic Adenocarcinoma Patients Treated with FOLFIRINOX. Clin Cancer Res 2018; 24:6204-6211. [DOI: 10.1158/1078-0432.ccr-18-1472] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/27/2018] [Accepted: 08/16/2018] [Indexed: 11/16/2022]
|
158
|
Brand R, Borazanci E, Speare V, Dudley B, Karloski E, Peters MLB, Stobie L, Bahary N, Zeh H, Zureikat A, Hogg M, Lee K, Tsung A, Rhee J, Ohr J, Sun W, Lee J, Moser AJ, DeLeonardis K, Krejdovsky J, Dalton E, LaDuca H, Dolinsky J, Colvin A, Lim C, Black MH, Tung N. Prospective study of germline genetic testing in incident cases of pancreatic adenocarcinoma. Cancer 2018; 124:3520-3527. [PMID: 30067863 DOI: 10.1002/cncr.31628] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/07/2018] [Accepted: 05/29/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND The objective of this study was to investigate the prevalence of pathogenic germline variants (PGVs) in 32 cancer susceptibility genes in individuals with newly diagnosed pancreatic ductal adenocarcinoma (PDAC). A key secondary objective was to evaluate how often PGVs would have been undetected with existing genetic testing criteria. METHODS From May 2016 through May 2017, this multicenter cohort study enrolled consecutive patients aged 18 to 89 years with histologically confirmed PDAC diagnosed within the previous 12 weeks. Demographics, medical histories, and 3-generation pedigrees were collected from participants who provided samples for germline DNA analysis. RESULTS Four hundred nineteen patients were deemed eligible, 302 were enrolled, and 298 were included in the final cohort. Clinically actionable variants were reported in 29 PDAC patients (9.7%), with 23 (7.7%) having a PGV associated with an increased risk for PDAC. Six of 23 individuals (26%) with PDAC-associated gene mutations did not meet currently established genetic testing criteria. According to guideline-based genetic testing, only 11 of the 23 PGVs (48%) in known PDAC genes would have been detected. Six additional patients (2%) had PGVs associated with an increased risk for other cancers. CONCLUSIONS These findings support the significant prevalence of PGVs associated with PDAC and the limitations of current paradigms for selecting patients for genetic testing, and they thereby lend support for universal germline multigene genetic testing in this population.
Collapse
Affiliation(s)
- Randall Brand
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | | | - Beth Dudley
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Eve Karloski
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Lindsey Stobie
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Nathan Bahary
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Herbert Zeh
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Amer Zureikat
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Melissa Hogg
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Kenneth Lee
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - John Rhee
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - James Ohr
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas, Kansas City, Kansas
| | - James Lee
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - A James Moser
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | - Nadine Tung
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
159
|
Hu C, LaDuca H, Shimelis H, Polley EC, Lilyquist J, Hart SN, Na J, Thomas A, Lee KY, Davis BT, Black MH, Pesaran T, Goldgar DE, Dolinsky JS, Couch FJ. Multigene Hereditary Cancer Panels Reveal High-Risk Pancreatic Cancer Susceptibility Genes. JCO Precis Oncol 2018; 2:PO.17.00291. [PMID: 31497750 PMCID: PMC6731034 DOI: 10.1200/po.17.00291] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The relevance of inherited pathogenic mutations in cancer predisposition genes in pancreatic cancer is not well understood. We aimed to assess the characteristics of patients with pancreatic cancer referred for hereditary cancer genetic testing and to estimate the risk of pancreatic cancer associated with mutations in panel-based cancer predisposition genes in this high-risk population. METHODS Patients with pancreatic cancer (N = 1,652) were identified from a 140,000-patient cohort undergoing multigene panel testing of predisposition genes between March 2012 and June 2016. Gene-level mutation frequencies relative to Exome Aggregation Consortium and Genome Aggregation Database reference controls were assessed. RESULTS The frequency of germline cancer predisposition gene mutations among patients with pancreatic cancer was 20.73%. Mutations in ATM, BRCA2, CDKN2A, MSH2, MSH6, PALB2, and TP53 were associated with high pancreatic cancer risk (odds ratio, > 5), and mutations in BRCA1 were associated with moderate risk (odds ratio, > 2). In a logistic regression model adjusted for age at diagnosis and family history of cancer, ATM and BRCA2 mutations were associated with personal history of breast or pancreatic cancer, whereas PALB2 mutations were associated with family history of breast or pancreatic cancer. CONCLUSION These findings provide insight into the spectrum of mutations expected in patients with pancreatic cancer referred for cancer predisposition testing. Mutations in eight genes confer high or moderate risk of pancreatic cancer and may prove useful for risk assessment for pancreatic and other cancers. Family and personal histories of breast cancer are strong predictors of germline mutations.
Collapse
Affiliation(s)
- Chunling Hu
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Holly LaDuca
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Hermela Shimelis
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Eric C. Polley
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Jenna Lilyquist
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Steven N. Hart
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Jie Na
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Abigail Thomas
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Kun Y. Lee
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Brigette Tippin Davis
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Mary Helen Black
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Tina Pesaran
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - David E. Goldgar
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Jill S. Dolinsky
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| | - Fergus J. Couch
- Chunling Hu, Hermela Shimelis, Eric C. Polley, Jenna Lilyquist, Steven N. Hart, Jie Na, Abigail Thomas, Kun Y. Lee, and Fergus J. Couch, Mayo Clinic, Rochester, MN; Holly LaDuca, Brigette Tippin Davis, Mary Helen Black, Tina Pesaran, and Jill S. Dolinsky, Ambry Genetics, Aliso Viejo, CA; and David E. Goldgar, University of Utah, Salt Lake City, UT
| |
Collapse
|
160
|
Hu C, Hart SN, Polley EC, Gnanaolivu R, Shimelis H, Lee KY, Lilyquist J, Na J, Moore R, Antwi SO, Bamlet WR, Chaffee KG, DiCarlo J, Wu Z, Samara R, Kasi PM, McWilliams RR, Petersen GM, Couch FJ. Association Between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. JAMA 2018; 319:2401-2409. [PMID: 29922827 PMCID: PMC6092184 DOI: 10.1001/jama.2018.6228] [Citation(s) in RCA: 356] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IMPORTANCE Individuals genetically predisposed to pancreatic cancer may benefit from early detection. Genes that predispose to pancreatic cancer and the risks of pancreatic cancer associated with mutations in these genes are not well defined. OBJECTIVE To determine whether inherited germline mutations in cancer predisposition genes are associated with increased risks of pancreatic cancer. DESIGN, SETTING, AND PARTICIPANTS Case-control analysis to identify pancreatic cancer predisposition genes; longitudinal analysis of patients with pancreatic cancer for prognosis. The study included 3030 adults diagnosed as having pancreatic cancer and enrolled in a Mayo Clinic registry between October 12, 2000, and March 31, 2016, with last follow-up on June 22, 2017. Reference controls were 123 136 individuals with exome sequence data in the public Genome Aggregation Database and 53 105 in the Exome Aggregation Consortium database. EXPOSURES Individuals were classified based on carrying a deleterious mutation in cancer predisposition genes and having a personal or family history of cancer. MAIN OUTCOMES AND MEASURES Germline mutations in coding regions of 21 cancer predisposition genes were identified by sequencing of products from a custom multiplex polymerase chain reaction-based panel; associations of genes with pancreatic cancer were assessed by comparing frequency of mutations in genes of pancreatic cancer patients with those of reference controls. RESULTS Comparing 3030 case patients with pancreatic cancer (43.2% female; 95.6% non-Hispanic white; mean age at diagnosis, 65.3 [SD, 10.7] years) with reference controls, significant associations were observed between pancreatic cancer and mutations in CDKN2A (0.3% of cases and 0.02% of controls; odds ratio [OR], 12.33; 95% CI, 5.43-25.61); TP53 (0.2% of cases and 0.02% of controls; OR, 6.70; 95% CI, 2.52-14.95); MLH1 (0.13% of cases and 0.02% of controls; OR, 6.66; 95% CI, 1.94-17.53); BRCA2 (1.9% of cases and 0.3% of controls; OR, 6.20; 95% CI, 4.62-8.17); ATM (2.3% of cases and 0.37% of controls; OR, 5.71; 95% CI, 4.38-7.33); and BRCA1 (0.6% of cases and 0.2% of controls; OR, 2.58; 95% CI, 1.54-4.05). CONCLUSIONS AND RELEVANCE In this case-control study, mutations in 6 genes associated with pancreatic cancer were found in 5.5% of all pancreatic cancer patients, including 7.9% of patients with a family history of pancreatic cancer and 5.2% of patients without a family history of pancreatic cancer. Further research is needed for replication in other populations.
Collapse
Affiliation(s)
- Chunling Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Steven N Hart
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Eric C Polley
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Rohan Gnanaolivu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Hermela Shimelis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kun Y Lee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jenna Lilyquist
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Jie Na
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Raymond Moore
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Samuel O Antwi
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - John DiCarlo
- Qiagen Sciences Research and Development, Qiagen Inc, Hilden, Germany
| | - Zhong Wu
- Qiagen Sciences Research and Development, Qiagen Inc, Hilden, Germany
| | - Raed Samara
- Qiagen Sciences Research and Development, Qiagen Inc, Hilden, Germany
| | | | | | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
161
|
Risk of multiple pancreatic cancers in CDKN2A-p16-Leiden mutation carriers. Eur J Hum Genet 2018; 26:1227-1229. [PMID: 29769629 DOI: 10.1038/s41431-018-0170-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 01/06/2023] Open
Abstract
CDKN2A-p16-Leiden mutation carriers have a substantial risk of developing pancreatic ductal adenocarcinoma (PDAC). One of the main clinical features of hereditary cancer is the development of multiple cancers. Since 2000, we have run a surveillance program for CDKN2A-p16-Leiden mutation carriers. The patients are offered a yearly MRI with optionally endoscopic ultrasound. In patients with a confirmed lesion, usually, a partial resection of the pancreas is recommended. A total of 18 PDAC (8.3%) were detected in 218 mutation carriers. In this report, we describe two CDKN2A-p16-Leiden patients with a synchronous and metachronous PDAC. Including two previously-reported cases, we identified four patients with multiple PDAC: two of 18 patients within the surveillance program (11%) and two patients with a proven CDKN2A-p16-Leiden mutation not participating in the surveillance program. In conclusion, this study demonstrated a high risk of developing multiple PDAC in CDKN2A-p16-Leiden mutation carriers. After detecting a primary tumor, it is very important to exclude the presence of a second synchronous tumor. Moreover, after a partial pancreatectomy for PDAC, close surveillance is necessary. In view of the current findings, offering a total pancreatectomy might be an appropriate option in patients with an early PDAC.
Collapse
|
162
|
NIPS, a 3D network-integrated predictor of deleterious protein SAPs, and its application in cancer prognosis. Sci Rep 2018; 8:6021. [PMID: 29662108 PMCID: PMC5902451 DOI: 10.1038/s41598-018-24286-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 03/27/2018] [Indexed: 12/20/2022] Open
Abstract
Identifying deleterious mutations remains a challenge in cancer genome sequencing projects, reflecting the vast number of candidate mutations per tumour and the existence of interpatient heterogeneity. Based on a 3D protein interaction network profiled via large-scale cross-linking mass spectrometry, we propose a weighted average formula involving the combination of three types of information into a 'meta-score'. We assume that a single amino acid polymorphism (SAP) may have a deleterious effect if the mutation rarely occurs naturally during evolution, if it inhibits binding between a pair of interacting proteins when located at their interface, or if it plays an important role in a protein interaction (PPI) network. Cross-validation indicated that this new method presents an AUC value of 0.93 and outperforms other widely used tools. The application of this method to the CPTAC colorectal cancer dataset enabled the accurate identification of validated deleterious mutations and yielded insights into their potential pathogenesis. Survival analysis showed that the accumulation of deleterious SAPs is significantly associated with a poor prognosis. The new method provides an alternative method to identifying and ranking deleterious cancer SAPs based on a 3D PPI network and will contribute to the understanding of pathogenesis and the discovery of prognostic biomarkers.
Collapse
|
163
|
Dudley B, Karloski E, Monzon FA, Singhi AD, Lincoln SE, Bahary N, Brand RE. Germline mutation prevalence in individuals with pancreatic cancer and a history of previous malignancy. Cancer 2018; 124:1691-1700. [PMID: 29360161 DOI: 10.1002/cncr.31242] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/22/2017] [Accepted: 12/29/2017] [Indexed: 12/17/2023]
Abstract
BACKGROUND Approximately 10% of pancreatic adenocarcinoma (PC) cases are attributed to hereditary causes. Individuals with PC and a personal history of another cancer associated with hereditary breast and ovarian cancer (HBOC) or Lynch syndrome (LS) may be more likely to carry germline mutations. METHODS Participants with PC and a history of cancer were selected from a pancreatic disease registry. Of 1296 individuals with PC, 149 had a relevant history of cancer. If banked DNA was available, a multigene panel was performed for individuals who had not 1) previously had a mutation identified through clinical testing or 2) undergone clinical multigene panel testing with no mutations detected. RESULTS Twenty-two of 124 individuals with PC and another HBOC- or LS-related cancer who underwent genetic testing had a mutation identified in a PC susceptibility gene (18%). If prostate cancer is excluded, the mutation prevalence increased to 23% (21/93). Mutation carriers were more likely to have more than 1 previous cancer diagnosis (P = .001), to have had clinical genetic testing (P = .001), and to meet National Comprehensive Cancer Network (NCCN) genetic testing criteria (P < .001). Approximately 23% of mutation carriers did not meet NCCN HBOC or LS testing guidelines based on their personal cancer history and reported cancer history in first-degree relatives. CONCLUSION At least 18% of individuals with PC and a personal history of other HBOC- or LS-related cancers carry mutations in a PC susceptibility gene based on our data, suggesting that criteria for genetic testing in individuals with PC should include consideration of previous cancer history. Cancer 2018;124:1691-700. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Beth Dudley
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eve Karloski
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Nathan Bahary
- Department of Hematology/Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Randall E Brand
- Department of Gastroenterology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|
164
|
Molina-Montes E, Gomez-Rubio P, Márquez M, Rava M, Löhr M, Michalski CW, Molero X, Farré A, Perea J, Greenhalf W, Ilzarbe L, O'Rorke M, Tardón A, Gress T, Barberà VM, Crnogorac-Jurcevic T, Domínguez-Muñoz E, Muñoz-Bellvís L, Balsells J, Costello E, Huang J, Iglesias M, Kleeff J, Kong B, Mora J, Murray L, O'Driscoll D, Poves I, Scarpa A, Ye W, Hidalgo M, Sharp L, Carrato A, Real FX, Malats N. Risk of pancreatic cancer associated with family history of cancer and other medical conditions by accounting for smoking among relatives. Int J Epidemiol 2018; 47:473-483. [PMID: 29329392 DOI: 10.1093/ije/dyx269] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2018] [Indexed: 12/16/2022] Open
Abstract
Background Family history (FH) of pancreatic cancer (PC) has been associated with an increased risk of PC, but little is known regarding the role of inherited/environmental factors or that of FH of other comorbidities in PC risk. We aimed to address these issues using multiple methodological approaches. Methods Case-control study including 1431 PC cases and 1090 controls and a reconstructed-cohort study (N = 16 747) made up of their first-degree relatives (FDR). Logistic regression was used to evaluate PC risk associated with FH of cancer, diabetes, allergies, asthma, cystic fibrosis and chronic pancreatitis by relative type and number of affected relatives, by smoking status and other potential effect modifiers, and by tumour stage and location. Familial aggregation of cancer was assessed within the cohort using Cox proportional hazard regression. Results FH of PC was associated with an increased PC risk [odds ratio (OR) = 2.68; 95% confidence interval (CI): 2.27-4.06] when compared with cancer-free FH, the risk being greater when ≥ 2 FDRs suffered PC (OR = 3.88; 95% CI: 2.96-9.73) and among current smokers (OR = 3.16; 95% CI: 2.56-5.78, interaction FHPC*smoking P-value = 0.04). PC cumulative risk by age 75 was 2.2% among FDRs of cases and 0.7% in those of controls [hazard ratio (HR) = 2.42; 95% CI: 2.16-2.71]. PC risk was significantly associated with FH of cancer (OR = 1.30; 95% CI: 1.13-1.54) and diabetes (OR = 1.24; 95% CI: 1.01-1.52), but not with FH of other diseases. Conclusions The concordant findings using both approaches strengthen the notion that FH of cancer, PC or diabetes confers a higher PC risk. Smoking notably increases PC risk associated with FH of PC. Further evaluation of these associations should be undertaken to guide PC prevention strategies.
Collapse
Affiliation(s)
- E Molina-Montes
- Spanish National Cancer Research Center (CNIO), Genetic and Molecular Epidemiology Group, Madrid, and CIBERONC, Spain
| | - P Gomez-Rubio
- Spanish National Cancer Research Center (CNIO), Genetic and Molecular Epidemiology Group, Madrid, and CIBERONC, Spain
| | - M Márquez
- Spanish National Cancer Research Center (CNIO), Genetic and Molecular Epidemiology Group, Madrid, and CIBERONC, Spain
| | - M Rava
- Spanish National Cancer Research Center (CNIO), Genetic and Molecular Epidemiology Group, Madrid, and CIBERONC, Spain
| | - M Löhr
- Karolinska Institutet and University Hospital, Gastrocentrum, Stockholm, Sweden
| | - C W Michalski
- Technical University of Munich, Department of Surgery, Munich, Germany
- University of Heidelberg, Department of Surgery, Heidelberg, Germany
| | - X Molero
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, and CIBEREHD, Spain
| | - A Farré
- Hospital de la Santa Creu i Sant Pau, Department of Gastroenterology, Barcelona, Spain
| | - J Perea
- University Hospital 12 de Octubre, Department of Surgery, Madrid, Spain
| | - W Greenhalf
- Royal Liverpool University Hospital, Department of Molecular and Clinical Cancer Medicine, Liverpool, UK
| | - L Ilzarbe
- Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - M O'Rorke
- Queen's University Belfast, Centre for Public Health, Belfast, UK
| | - A Tardón
- Instituto Universitario de Oncología del Principado de Asturias, Department of Medicine, Oviedo, and CIBERESP, Spain
| | - T Gress
- University Hospital of Giessen and Marburg, Department of Gastroenterology, Marburg, Germany
| | - V M Barberà
- General University Hospital of Elche, Molecular Genetics Laboratory, Elche, Spain
| | - T Crnogorac-Jurcevic
- Barts Cancer Institute, Centre for Molecular Oncology, Queen Mary University of London, London, UK
| | - E Domínguez-Muñoz
- University Clinical Hospital of Santiago de Compostela, Department of Gastroenterology, Santiago de Compostela, Spain
| | - L Muñoz-Bellvís
- Salamanca University Hospital, General and Digestive Surgery Department, Salamanca, Spain
| | - J Balsells
- Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, and CIBEREHD, Spain
| | - E Costello
- Royal Liverpool University Hospital, Department of Molecular and Clinical Cancer Medicine, Liverpool, UK
| | - J Huang
- Karolinska Institutet and University Hospital, Gastrocentrum, Stockholm, Sweden
| | - M Iglesias
- Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - J Kleeff
- Technical University of Munich, Department of Surgery, Munich, Germany
- Martin-Luther-University Halle-Wittenberg, Department of Visceral, Vascular and Endocrine Surgery, Halle (Saale), Germany
| | - Bo Kong
- Technical University of Munich, Department of Surgery, Munich, Germany
| | - J Mora
- Hospital de la Santa Creu i Sant Pau, Department of Gastroenterology, Barcelona, Spain
| | - L Murray
- Queen's University Belfast, Centre for Public Health, Belfast, UK
| | - D O'Driscoll
- National Cancer Registry Ireland and HRB Clinical Research Facility, University College Cork, Cork, Ireland
| | - I Poves
- Hospital del Mar-Parc de Salut Mar, Barcelona, Spain
| | - A Scarpa
- ARC-Net Centre for Applied Research on Cancer and Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - W Ye
- Karolinska Institutet and University Hospital, Gastrocentrum, Stockholm, Sweden
| | - M Hidalgo
- Madrid-Norte-Sanchinarro Hospital, Madrid, Spain
| | - L Sharp
- National Cancer Registry Ireland and HRB Clinical Research Facility, University College Cork, Cork, Ireland
- Newcastle University, Institute of Health and Society, Newcastle upon Tyne, UK
| | - A Carrato
- Ramón y Cajal University Hospital, Department of Oncology, IRYCIS, Alcala University, Madrid, and CIBERONC, Spain
| | - F X Real
- Spanish National Cancer Research Centre (CNIO), Epithelial Carcinogenesis Group, Madrid, Universitat Pompeu Fabra, Departament de Ciències Experimentals i de la Salut, Barcelona, and CIBERONC, Spain
| | - N Malats
- Spanish National Cancer Research Center (CNIO), Genetic and Molecular Epidemiology Group, Madrid, and CIBERONC, Spain
| |
Collapse
|
165
|
Underhill M, Hong F, Lawrence J, Blonquist T, Syngal S. Relationship between individual and family characteristics and psychosocial factors in persons with familial pancreatic cancer. Psychooncology 2018; 27:1711-1718. [PMID: 29570238 DOI: 10.1002/pon.4712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 02/27/2018] [Accepted: 03/09/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Describe relationships between self-reported personal demographics or familial characteristics and psychosocial outcomes (Patient Reported Outcome Measurement Information System Global Health, Impact of Event Scale-Revised [pancreatic cancer risk-related distress], cancer risk perception, and cancer worry) in participants with inherited or familial pancreatic cancer risk. METHODS A multisite cross sectional survey of adults with elevated pancreatic cancer risk based on family history. All variables were summarized with descriptive statistics. To assess univariate associations, t test and chi-square/Fisher's exact test were used, and backward model selection was used in multivariable analysis. RESULTS Respondents (N = 132) reported moderate to high frequency of cancer worry and 59.3% perceived a 50% or more perceived lifetime risk for pancreatic cancer, which far exceeds objective risk estimates. Cancer worry was associated with female gender (P = .03) and pancreatic cancer risk specific distress (P = .05). Higher-risk perception was associated with having a high school education or less (P = .001), higher distress (P = .02), and cancer worry (P = .008) and family cancer death experience (P = .02). Higher distress was associated with experience as a caregiver to a seriously ill family member in the past 5 years (P = .006). CONCLUSIONS Individuals with inherited or familial pancreatic cancer risk experience cancer worry, distress, and have increased risk perception, particularly in the period following caring for a loved one with cancer. Routine evaluation of distress in this setting, as well as the development of supportive care resources, will help support patients living with risk for pancreatic cancer.
Collapse
Affiliation(s)
- Meghan Underhill
- The Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Fangxin Hong
- Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Janette Lawrence
- Center for Cancer Risk Assessment, Massachusetts General Hospital, Boston, MA, USA
| | - Traci Blonquist
- Biostatistics & Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sapna Syngal
- GI Cancer Genetics and Prevention Program, Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
166
|
Cremin C, Howard S, Le L, Karsan A, Schaeffer DF, Renouf D, Schrader KA. CDKN2A founder mutation in pancreatic ductal adenocarcinoma patients without cutaneous features of Familial Atypical Multiple Mole Melanoma (FAMMM) syndrome. Hered Cancer Clin Pract 2018. [PMID: 29541281 PMCID: PMC5842519 DOI: 10.1186/s13053-018-0088-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Approximately 5% to 10% of pancreatic ductal adenocarcinoma (PDAC) has a hereditary basis. In most of these defined hereditary cancer syndromes, PDAC is not the predominant cancer type. Traditional criteria for publicly funded genetic testing typically require the presence of a set combination of the predominant syndrome-associated cancer types in the family history. We report the identification of a CDKN2A pathogenic variant in a PDAC-prone family without the cutaneous features of multiple moles or melanoma that are characteristic of the Familial Atypical Multiple Mole Melanoma (FAMMM) Syndrome identified in a universal testing algorithm for inherited mutations in pancreatic cancer patients. Case presentation We present the case of two brothers of English ancestry diagnosed with PDAC within the same 12 month period, at the respective ages of 63 and 64 years of age. Neither brother reported a personal history of multiple moles or melanoma. Family history was positive for two second-degree relatives diagnosed with PDAC but was negative for other cancers or multiple moles in first- and second-degree relatives. Due to the absence of melanoma, this family did not meet provincial criteria for publicly funded genetic testing. Clinical genetic testing offered through a research grant identified a pathogenic variant in the CDKN2A gene c.377 T > A (p.Val126Asp). This variant is a North American founder mutation believed to pre-date colonization. Conclusions This case reminds clinicians to consider the possibility of a germline CDKN2A mutation in families with a high prevalence of PDAC, even in the absence of moles or melanoma. This case supports recent guidelines published by the American College of Medical Genetics and Genomics (ACMG) that genetics referrals are indicated in families with three or more cases of PDAC regardless of other cancer types in the family. A multi-gene panel approach is of particular benefit in diagnosing inherited cancer susceptibility in PDAC-only families.
Collapse
Affiliation(s)
- Carol Cremin
- 1Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia Canada.,Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia Canada.,Pancreas Centre BC, Vancouver, British Columbia Canada
| | - Sarah Howard
- Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia Canada.,Pancreas Centre BC, Vancouver, British Columbia Canada
| | - Lyly Le
- Department of Medical Oncology, BC Cancer - Surrey, Surrey, British Columbia Canada
| | - Aly Karsan
- 4Centre for Clinical Genomics, Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia Canada.,5Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - David F Schaeffer
- Pancreas Centre BC, Vancouver, British Columbia Canada.,7Division of Anatomical Pathology, Vancouver General Hospital, Vancouver, British Columbia Canada
| | - Daniel Renouf
- Pancreas Centre BC, Vancouver, British Columbia Canada.,Division of Medical Oncology, BC Cancer, Vancouver, British Columbia Canada
| | - Kasmintan A Schrader
- 1Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia Canada.,Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia Canada.,Pancreas Centre BC, Vancouver, British Columbia Canada.,9Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia Canada
| |
Collapse
|
167
|
Inherited DNA-Repair Defects in Colorectal Cancer. Am J Hum Genet 2018; 102:401-414. [PMID: 29478780 DOI: 10.1016/j.ajhg.2018.01.018] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) heritability has been estimated to be around 30%. However, mutations in the known CRC-susceptibility genes explain CRC risk in fewer than 10% of affected individuals. Germline mutations in DNA-repair genes (DRGs) have recently been reported in CRC, but their contribution to CRC risk is largely unknown. We evaluated the gene-level germline mutation enrichment of 40 DRGs in 680 unselected CRC individuals and 27,728 ancestry-matched cancer-free adults. Significant findings were then examined in independent cohorts of 1,661 unselected CRC individuals and 1,456 individuals with early-onset CRC. Of the 680 individuals in the discovery set, 31 (4.56%) individuals harbored germline pathogenic mutations in known CRC-susceptibility genes, and another 33 (4.85%) individuals had DRG mutations that have not been previously associated with CRC risk. Germline pathogenic mutations in ATM and PALB2 were enriched in both the discovery (OR = 2.81 and p = 0.035 for ATM and OR = 4.91 and p = 0.024 for PALB2) and validation (OR = 2.97 and adjusted p = 0.0013 for ATM and OR = 3.42 and adjusted p = 0.034 for PALB2) sets. Biallelic loss of ATM was evident in all individuals with matched tumor profiling. CRC individuals also had higher rates of actionable mutations in the HR pathway, which can substantially increase the risk of developing cancers other than CRC. Our analysis provides evidence for ATM and PALB2 as CRC-risk genes, underscoring the importance of the homologous recombination pathway in CRC. In addition, we identified frequent complete homologous recombination deficiency in CRC tumors, representing a unique opportunity to explore targeted therapeutic interventions such as poly-ADP ribose polymerase inhibitor (PARPi).
Collapse
|
168
|
Blair AB, Groot VP, Gemenetzis G, Wei J, Cameron JL, Weiss MJ, Goggins M, Wolfgang CL, Yu J, He J. BRCA1/BRCA2 Germline Mutation Carriers and Sporadic Pancreatic Ductal Adenocarcinoma. J Am Coll Surg 2018; 226:630-637.e1. [PMID: 29309945 DOI: 10.1016/j.jamcollsurg.2017.12.021] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 12/18/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The outcomes of sporadic pancreatic ductal adenocarcinoma (PDAC) patients with germline mutations of BRCA1/BRCA2 remains unclear. The prognostic significance of BRCA1/BRCA2 mutations on survival is not well established. STUDY DESIGN We performed targeted next-generation sequencing (NGS) to identify BRCA1/BRCA2 germline mutations in resected sporadic PDAC cases from 2000 to 2015. Germline BRCA mutation carriers were matched by age and tumor location to those with BRCA1/BRCA2 wild-type genes from our institutional database. Demographics, clinicopathologic features, overall survival (OS), and disease-free survival (DFS) were abstracted from medical records and compared between the 2 cohorts. RESULTS Twenty-two patients with sporadic cancer and BRCA1 (n = 4) or BRCA2 (n = 18) germline mutations and 105 wild-type patients were identified for this case-control study. The BRCA1/BRCA2 mutations were associated with inferior median OS (20.2 vs 27.8 months, p = 0.034) and DFS (8.4 vs 16.7 months, p < 0.001) when compared with the matched wild-type controls. On multivariable analyses, a BRCA1/BRCA2 mutation (hazard ratio [HR] 2.10, p < 0.001), positive margin status (HR 1.72, p = 0.021), and lack of adjuvant therapy (HR 2.38, p < 0.001), were all independently associated with worse survival. Within the BRCA1/BRCA2 mutated group, having had platinum-based adjuvant chemotherapy (n = 10) was associated with better survival than alternative chemotherapy (n = 8) or no adjuvant therapy (n = 4) (31.0 vs 17.8 vs 9.3 months, respectively, p < 0.001). CONCLUSIONS Carriers of BRCA1/BRCA2 mutation with sporadic PDAC had a worse survival after pancreatectomy than their BRCA wild-type counterparts. However, platinum-based chemotherapy regimens were associated with markedly improved survival in patients with BRCA1/BRCA2 mutations, with survival differences no longer appreciated with wild-type patients.
Collapse
Affiliation(s)
- Alex B Blair
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Vincent P Groot
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Georgios Gemenetzis
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jishu Wei
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - John L Cameron
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Matthew J Weiss
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Christopher L Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jun Yu
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jin He
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, MD.
| |
Collapse
|
169
|
Lowery MA, Kelsen DP, Capanu M, Smith SC, Lee JW, Stadler ZK, Moore MJ, Kindler HL, Golan T, Segal A, Maynard H, Hollywood E, Moynahan M, Salo-Mullen EE, Do RKG, Chen AP, Yu KH, Tang LH, O'Reilly EM. Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. Eur J Cancer 2017; 89:19-26. [PMID: 29223478 DOI: 10.1016/j.ejca.2017.11.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 10/24/2017] [Accepted: 11/01/2017] [Indexed: 02/09/2023]
Abstract
PURPOSE BRCA-associated cancers have increased sensitivity to poly(ADP-ribose) polymerase inhibitors (PARPis). This single arm, non-randomised, multicentre phase II trial evaluated the response rate of veliparib in patients with previously treated BRCA1/2- or PALB2-mutant pancreatic adenocarcinoma (PDAC). METHODS Patients with stage III/IV PDAC and known germline BRCA1/2 or PALB2 mutation, 1-2 lines of treatment, Eastern Cooperative Oncology Group 0-2, were enrolled. Veliparib was dosed at a volume of 300 mg twice-daily (N = 3), then 400 mg twice-daily (N = 15) days 1-28. The primary end-point was to determine the response rate of veliparib; secondary end-points included progression-free survival (PFS), duration of response, overall survival (OS) and safety. RESULTS Sixteen patients were enrolled; male N = 8 (50%). Median age was 52 years (range 43-77). Five (31%) had a BRCA1 and 11 (69%) had a BRCA2 mutation. Fourteen (88%) patients had received prior platinum-based therapy. No confirmed partial responses (PRs) were seen: one (6%) unconfirmed PR was observed at 4 months with disease progression (PD) at 6 months; four (25%) had stable disease (SD), whereas 11 (69%) had PD as best response including one with clinical PD. Median PFS was 1.7 months (95% confidence interval [CI] 1.57-1.83) and median OS was 3.1 months (95% CI 1.9-4.1). Six (38%) patients had grade III toxicity, including fatigue (N = 3), haematology (N = 2) and nausea (N = 1). CONCLUSIONS Veliparib was well tolerated, but no confirmed response was observed although four (25%) patients remained on study with SD for ≥ 4 months. Additional strategies in this population are needed, and ongoing trials are evaluating PARPis combined with chemotherapy (NCT01585805) and as a maintenance strategy (NCT02184195).
Collapse
Affiliation(s)
- Maeve A Lowery
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - David P Kelsen
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | | | - Sloane C Smith
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan W Lee
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zsofia K Stadler
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Malcolm J Moore
- Princess Margaret Cancer Center- University Health Network, Toronto, Canada
| | | | - Talia Golan
- Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amiel Segal
- Share Zedek Medical Center, Jerusalem, Israel
| | - Hannah Maynard
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - MaryEllen Moynahan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | | | | | | | - Kenneth H Yu
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Laura H Tang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eileen M O'Reilly
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
170
|
Muñoz AR, Chakravarthy D, Gong J, Halff GA, Ghosh R, Kumar AP. Pancreatic cancer: Current status and Challenges. CURRENT PHARMACOLOGY REPORTS 2017; 3:396-408. [PMID: 29404265 PMCID: PMC5795623 DOI: 10.1007/s40495-017-0112-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF THE REVIEW The 5-year survival rate of patients with pancreatic cancer (PanCA) has remained stagnant. Unfortunately, the incidence is almost equal to mortality rates. These facts underscore the importance of concerted efforts to understand the pathology of this disease. Deregulation of multiple signaling pathways involved in a wide variety of cellular processes including proliferation, apoptosis, invasion, and metastasis contribute not only to cancer development but also to therapeutic resistance. The purpose of this review is to summarize current understanding of etiological factors including emerging evidence on the role of infectious agents, factors associated with therapeutic resistance and therapeutic options. RECENT FINDINGS The unique aspect of PanCA is "desmoplasia", a process that involves proliferation of stromal fibroblasts and collagen deposition in and around the filtrating cancer. Recent studies have identified pancreatic stellate cells (PSCs) as a potential source of such desmoplasia. Biphasic interactions between PSCs and cancer cells, endothelial cells, and/or myeloid derived suppressor cells in the tumor microenvironment contribute to pancreatic carcinogenesis. SUMMARY We summarize limitations of current therapeutic approaches and potential strategies to overcome these limitations using natural products including botanicals as adjuvant/neo-adjuvant for effective management of PanCA.
Collapse
Affiliation(s)
- Amanda R Muñoz
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX
| | | | | | - Glenn A Halff
- Department of Surgery, The University of Texas Health Science Center, San Antonio, TX
| | - Rita Ghosh
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX
- Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, TX
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX
- UT Health San Antonio Cancer Center, The University of Texas Health Science Center, San Antonio, TX
| | - Addanki P Kumar
- Department of Urology, The University of Texas Health Science Center, San Antonio, TX
- Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, TX
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX
- UT Health San Antonio Cancer Center, The University of Texas Health Science Center, San Antonio, TX
- South Texas Veterans Health Care System, The University of Texas Health Science Center, San Antonio, TX
| |
Collapse
|
171
|
Abstract
Pancreatic cancers arise through a series of genetic events both inherited and acquired. Inherited genetic changes, both high penetrance and low penetrance, are an important component of pancreatic cancer risk, and may be used to characterize populations who will benefit from early detection. Furthermore, pancreatic cancer patients with inherited mutations may be particularly sensitive to certain targeted agents, providing an opportunity to personalized treatment. Family history of pancreatic cancer is one of the strongest risk factors for the disease, and is associated with an increased risk of caners at other sites, including but not limited to breast, ovarian and colorectal cancer. The goal of this chapter is to discuss the importance of family history of pancreatic cancer, and the known genes that account for a portion of the familial clustering of pancreatic cancer.
Collapse
Affiliation(s)
- Fei Chen
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicholas J Roberts
- Department of Pathology, Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institution, Baltimore, MD, USA
| | - Alison P Klein
- Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Department of Pathology, Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institution, Baltimore, MD, USA.
| |
Collapse
|
172
|
Goehringer C, Sutter C, Kloor M, Gebert J, Slater EP, Keller M, Treiber I, Ganschow P, Kadmon M, Moog U. Double germline mutations in APC and BRCA2 in an individual with a pancreatic tumor. Fam Cancer 2017; 16:303-309. [PMID: 27838800 DOI: 10.1007/s10689-016-9952-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We report on three brothers affected by pancreatic tumors, all due to different causes, including mutations associated with two different cancer predisposition syndromes in the same individual. In the index patient a germline mutation both in the APC and BRCA2 gene was identified while one affected brother showed the BRCA2 mutation only and another brother is supposed to have developed pancreatic cancer due to multiple non-genetic risk factors. We outline the impact of a double germline mutation in two tumor predisposition genes in one individual and proven heterogeneity of multiple cases of pancreatic tumors in one family. With the growing implementation of next generation sequence based panel testing for multiple genes involved in tumor predisposition syndromes, relevant variants in two (or more) genes will be found more frequently. This family illustrates the importance of family studies, especially when using gene panel tests.
Collapse
Affiliation(s)
- Caroline Goehringer
- Institute of Human Genetics, Heidelberg University, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany.
| | - Christian Sutter
- Institute of Human Genetics, Heidelberg University, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 220/221, 69120, Heidelberg, Germany
| | - Johannes Gebert
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University, Im Neuenheimer Feld 220/221, 69120, Heidelberg, Germany
| | - Emily P Slater
- Department of Surgery, Philipps-University Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Monika Keller
- Department of Psychosomatic and General Clinical Medicine, Heidelberg University, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Irmgard Treiber
- Department of General Surgery, Heidelberg University, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Petra Ganschow
- Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery, Hospital of the University of Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Martina Kadmon
- Faculty of Medicine and Health Sciences, University of Oldenburg, Carl-von-Ossietzky-Str. 9-11, 26129, Oldenburg, Germany
| | - Ute Moog
- Institute of Human Genetics, Heidelberg University, Im Neuenheimer Feld 440, 69120, Heidelberg, Germany
| |
Collapse
|
173
|
Shindo K, Yu J, Suenaga M, Fesharakizadeh S, Cho C, Macgregor-Das A, Siddiqui A, Witmer PD, Tamura K, Song TJ, Navarro Almario JA, Brant A, Borges M, Ford M, Barkley T, He J, Weiss MJ, Wolfgang CL, Roberts NJ, Hruban RH, Klein AP, Goggins M. Deleterious Germline Mutations in Patients With Apparently Sporadic Pancreatic Adenocarcinoma. J Clin Oncol 2017; 35:3382-3390. [PMID: 28767289 DOI: 10.1200/jco.2017.72.3502] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose Deleterious germline mutations contribute to pancreatic cancer susceptibility and are well documented in families in which multiple members have had pancreatic cancer. Methods To define the prevalence of these germline mutations in patients with apparently sporadic pancreatic cancer, we sequenced 32 genes, including known pancreatic cancer susceptibility genes, in DNA prepared from normal tissue obtained from 854 patients with pancreatic ductal adenocarcinoma, 288 patients with other pancreatic and periampullary neoplasms, and 51 patients with non-neoplastic diseases who underwent pancreatic resection at Johns Hopkins Hospital between 2000 and 2015. Results Thirty-three (3.9%; 95% CI, 3.0% to 5.8%) of 854 patients with pancreatic cancer had a deleterious germline mutation, 31 (3.5%) of which affected known familial pancreatic cancer susceptibility genes: BRCA2 (12 patients), ATM (10 patients), BRCA1 (3 patients), PALB2 (2 patients), MLH1 (2 patients), CDKN2A (1 patient), and TP53 (1 patient). Patients with these germline mutations were younger than those without (mean ± SD, 60.8 ± 10.6 v 65.1 ± 10.5 years; P = .03). Deleterious germline mutations were also found in BUB1B (1) and BUB3 (1). Only three of these 33 patients had reported a family history of pancreatic cancer, and most did not have a cancer family history to suggest an inherited cancer syndrome. Five (1.7%) of 288 patients with other periampullary neoplasms also had a deleterious germline mutation. Conclusion Germline mutations in pancreatic cancer susceptibility genes are commonly identified in patients with pancreatic cancer without a significant family history of cancer. These deleterious pancreatic cancer susceptibility gene mutations, some of which are therapeutically targetable, will be missed if current family history guidelines are the main criteria used to determine the appropriateness of gene testing.
Collapse
Affiliation(s)
- Koji Shindo
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Jun Yu
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Masaya Suenaga
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Shahriar Fesharakizadeh
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Christy Cho
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Anne Macgregor-Das
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Abdulrehman Siddiqui
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - P Dane Witmer
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Koji Tamura
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Tae Jun Song
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | | | - Aaron Brant
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Michael Borges
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Madeline Ford
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Thomas Barkley
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Jin He
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Matthew J Weiss
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Christopher L Wolfgang
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Nicholas J Roberts
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Ralph H Hruban
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Alison P Klein
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| | - Michael Goggins
- All authors: The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
174
|
Chaffee KG, Oberg AL, McWilliams RR, Majithia N, Allen BA, Kidd J, Singh N, Hartman AR, Wenstrup RJ, Petersen GM. Prevalence of germ-line mutations in cancer genes among pancreatic cancer patients with a positive family history. Genet Med 2017; 20:119-127. [PMID: 28726808 PMCID: PMC5760284 DOI: 10.1038/gim.2017.85] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 05/01/2017] [Indexed: 12/11/2022] Open
Abstract
Background Panel-based genetic testing has identified increasing numbers of patients with pancreatic ductal adenocarcinoma (PDAC) who carry germline mutations. However, small sample sizes or number of genes evaluated limit prevalence estimates of these mutations. We estimated prevalence of mutations in PDAC patients with positive family history. Methods We sequenced 25 cancer susceptibility genes in lymphocyte DNA from 302 PDAC patients in the Mayo Clinic Biospecimen Resource for Pancreatic Research Registry. Kindreds containing at least two first-degree relatives with PDAC met criteria for Familial Pancreatic Cancer (FPC), while the remaining were familial, but not FPC. Results Thirty-six patients (12%) carried at least one deleterious mutation in one of 11 genes. Of FPC patients, 25/185 (14%) were carriers, while 11/117 (9%) non-FPC patients with family history were carriers. Deleterious mutations (n) identified in PDAC patients were BRCA2 (11), ATM (8), CDKN2A (4), CHEK2 (4), MUTYH/MYH (3 heterozygotes, not biallelic), BRCA1 (2), and 1 each in BARD1, MSH2, NBN, PALB2, and PMS2. Novel mutations were found in ATM, BARD1, and PMS2. Conclusions Multiple susceptibility gene testing in PDAC patients with family history of pancreatic cancer is warranted regardless of FPC status, and will inform genetic risk counseling for families.
Collapse
Affiliation(s)
- Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Neil Majithia
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Brian A Allen
- Myriad Genetics Laboratories, Inc., Salt Lake City, Utah, USA
| | - John Kidd
- Myriad Genetics Laboratories, Inc., Salt Lake City, Utah, USA
| | - Nanda Singh
- Myriad Genetics Laboratories, Inc., Salt Lake City, Utah, USA
| | | | | | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
175
|
Moutinho-Ribeiro P, Coelho R, Giovannini M, Macedo G. Pancreatic cancer screening: Still a delusion? Pancreatology 2017; 17:754-765. [PMID: 28739291 DOI: 10.1016/j.pan.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/17/2017] [Accepted: 07/05/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic adenocarcinoma represents the fourth most common cause of cancer mortality and death due to pancreatic cancer (PC) have increased since 2003. Its incidence has also raised about 30% in the past decade and it is expected to become the second cause of cancer mortality by 2020 in the USA. Most PC present with metastatic disease and improvements in treatment outcomes for this group have been disappointing. These observations support the idea that screening to identify patients at an earlier stage might be an important strategy in improving overall PC outcomes. Many protocols have been tested, nevertheless, by now there is no effective screening program. Given the overall low incidence of disease and the current lack of accurate, inexpensive and noninvasive screening tests, the consensus is that widespread population-based screening for PC in the general population or in patients with only one affected first-degree relative is neither practicable nor indicated in most countries. However, a different scenario is screening patients with higher risk for PC, most of them with hereditary conditions predisposing the development of this neoplasia. In fact, some guidelines are now available helping to select these individuals at risk and to screen them, in order to achieve early detection of PC.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal.
| | - Rosa Coelho
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
| | - Marc Giovannini
- Endoscopic Unit, Paoli-Calmettes Institute, Marseilles, France
| | - Guilherme Macedo
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|
176
|
Connor AA, Gallinger S. Next generation sequencing of pancreatic ductal adenocarcinoma: right or wrong? Expert Rev Gastroenterol Hepatol 2017; 11:683-694. [PMID: 28460572 DOI: 10.1080/17474124.2017.1324296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has the highest mortality rate of all epithelial malignancies and a paradoxically rising incidence rate. Clinical translation of next generation sequencing (NGS) of tumour and germline samples may ameliorate outcomes by identifying prognostic and predictive genomic and transcriptomic features in appreciable fractions of patients, facilitating enrolment in biomarker-matched trials. Areas covered: The literature on precision oncology is reviewed. It is found that outcomes may be improved across various malignancies, and it is suggested that current issues of adequate tissue acquisition, turnaround times, analytic expertise and clinical trial accessibility may lessen as experience accrues. Also reviewed are PDAC genomic and transcriptomic NGS studies, emphasizing discoveries of promising biomarkers, though these require validation, and the fraction of patients that will benefit from these outside of the research setting is currently unknown. Expert commentary: Clinical use of NGS with PDAC should be used in investigational contexts in centers with multidisciplinary expertise in cancer sequencing and pancreatic cancer management. Biomarker directed studies will improve our understanding of actionable genomic variation in PDAC, and improve outcomes for this challenging disease.
Collapse
Affiliation(s)
- Ashton A Connor
- a PanCuRx Translational Research Initiative , Ontario Institute for Cancer Research , Toronto , Ontario , Canada.,b Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Ontario , Canada.,c Hepatobiliary/Pancreatic Surgical Oncology Program , University Health Network , Toronto , Ontario , Canada
| | - Steven Gallinger
- a PanCuRx Translational Research Initiative , Ontario Institute for Cancer Research , Toronto , Ontario , Canada.,b Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Ontario , Canada.,c Hepatobiliary/Pancreatic Surgical Oncology Program , University Health Network , Toronto , Ontario , Canada
| |
Collapse
|
177
|
Carrera S, Sancho A, Azkona E, Azkuna J, Lopez-Vivanco G. Hereditary pancreatic cancer: related syndromes and clinical perspective. Hered Cancer Clin Pract 2017; 15:9. [PMID: 28670351 PMCID: PMC5490219 DOI: 10.1186/s13053-017-0069-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/21/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is a very aggressive disease with a poor prognosis. The majority of them are attributed to sporadic causes, especially to many modifiable risk factors such as tobacco or alcohol abuse. The principal histologic subtype of pancreatic cancer is ductal adenocarcinoma. Pancreatic neuroendocrine tumors, which constitute a more indolent entity, represent second type of pancreatic cancer in terms of incidence. Individuals with a family history of pancreatic cancer carry an increased risk of developing the disease, which may be related to an underlying hereditary component. Unfortunately, in the majority of these families the suspected germline genetic cause responsible of the disease will not be identified, but approximately in a 20% of the cases a hereditary cancer predisposition syndrome with increased risk of pancreatic cancer development can be recognized. This review will be focused on the leading hereditary cancer syndromes related to pancreatic ductal adenocarcinoma and pancreatic neuroendocrine tumors. Additionally, we will try to explain clinical aspects related to the identification of germline mutations in pancreatic cancer patients and their potential implications in oncologic treatment decisions.
Collapse
Affiliation(s)
- Sergio Carrera
- Hereditary Cancer Genetic Counseling Unit- Medical Oncology Department, Cruces University Hospital, Plaza de Cruces s/n. 48903, Baracaldo, Bizkaia Spain
| | - Aintzane Sancho
- Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | - Eider Azkona
- Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | - Josune Azkuna
- Medical Oncology Department, Cruces University Hospital, Baracaldo, Spain
| | | |
Collapse
|
178
|
Genomic Variations in Pancreatic Cancer and Potential Opportunities for Development of New Approaches for Diagnosis and Treatment. Int J Mol Sci 2017; 18:ijms18061201. [PMID: 28587243 PMCID: PMC5486024 DOI: 10.3390/ijms18061201] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/30/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Human pancreatic cancer has a very poor prognosis with an overall five-year survival rate of less than 5% and an average median survival time of six months. This is largely due to metastatic disease, which is already present in the majority of patients when diagnosed. Although our understanding of the molecular events underlying multi-step carcinogenesis in pancreatic cancer has steadily increased, translation into more effective therapeutic approaches has been inefficient in recent decades. Therefore, it is imperative that novel and targeted approaches are designed to facilitate the early detection and treatment of pancreatic cancer. Presently, there are numerous ongoing studies investigating the types of genomic variations in pancreatic cancer and their impact on tumor initiation and growth, as well as prognosis. This has led to the development of therapeutics to target these genetic variations for clinical benefit. Thus far, there have been minimal clinical successes directly targeting these genomic alterations; however research is ongoing to ultimately discover an innovative approach to tackle this devastating disease. This review will discuss the genomic variations in pancreatic cancer, and the resulting potential diagnostic and therapeutic implications.
Collapse
|
179
|
Diwakarla C, Hannan K, Hein N, Yip D. Advanced pancreatic ductal adenocarcinoma - Complexities of treatment and emerging therapeutic options. World J Gastroenterol 2017; 23:2276-2285. [PMID: 28428707 PMCID: PMC5385394 DOI: 10.3748/wjg.v23.i13.2276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/20/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a poor prognosis regardless of stage. To date the mainstay of therapy for advanced disease has been chemotherapy with little incremental improvements in outcome. Despite extensive research investigating new treatment options the current practices continue to utilise fluorouracil or gemcitabine containing combinations. The need for novel therapeutic approaches is mandated by the ongoing poor survival rates associated with this disease. One such approach may include manipulation of ribosome biogenesis and the nucleolar stress response, which has recently been applied to haematological malignancies such as lymphoma and prostate cancer with promising results. This review will focus on the current therapeutic options for pancreatic ductal adenocarcinoma and the complexities associated with developing novel treatments, with a particular emphasis on the role of the nucleolus as a treatment strategy.
Collapse
|
180
|
Dreyer SB, Chang DK, Bailey P, Biankin AV. Pancreatic Cancer Genomes: Implications for Clinical Management and Therapeutic Development. Clin Cancer Res 2017; 23:1638-1646. [PMID: 28373362 DOI: 10.1158/1078-0432.ccr-16-2411] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 01/25/2017] [Accepted: 01/27/2017] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer has become the third leading cause of cancer-related death, with little improvement in outcomes despite decades of research. Surgery remains the only chance of cure, yet only 20% of patients will be alive at 5 years after pancreatic resection. Few chemotherapeutics provide any improvement in outcome, and even then, for approved therapies, the survival benefits are marginal. Genomic sequencing studies of pancreatic cancer have revealed a small set of consistent mutations found in most pancreatic cancers and beyond that, a low prevalence for targetable mutations. This may explain the failure of conventional clinical trial designs to show any meaningful survival benefit, except in small and undefined patient subgroups. With the development of next-generation sequencing technology, genomic sequencing and analysis can be performed in a clinically meaningful turnaround time. This can identify therapeutic targets in individual patients and personalize treatment selection. Incorporating preclinical discovery and molecularly guided therapy into clinical trial design has the potential to significantly improve outcomes in this lethal malignancy. In this review, we discuss the findings of recent large-scale genomic sequencing projects in pancreatic cancer and the potential relevance of these data to therapeutic development. Clin Cancer Res; 23(7); 1638-46. ©2017 AACRSee all articles in this CCR Focus section, "Pancreatic Cancer: Challenge and Inspiration."
Collapse
Affiliation(s)
- Stephan B Dreyer
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, United Kingdom
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, United Kingdom
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Peter Bailey
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, United Kingdom
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Bearsden, Glasgow, Scotland, United Kingdom.
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, United Kingdom
- South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, New South Wales, Australia
| |
Collapse
|
181
|
Gravells P, Grant E, Smith KM, James DI, Bryant HE. Specific killing of DNA damage-response deficient cells with inhibitors of poly(ADP-ribose) glycohydrolase. DNA Repair (Amst) 2017; 52:81-91. [PMID: 28254358 PMCID: PMC5360195 DOI: 10.1016/j.dnarep.2017.02.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/16/2017] [Accepted: 02/13/2017] [Indexed: 12/12/2022]
Abstract
Poly(ADP-ribosylation) of proteins following DNA damage is well studied and the use of poly(ADP-ribose) polymerase (PARP) inhibitors as therapeutic agents is an exciting prospect for the treatment of many cancers. Poly(ADP-ribose) glycohydrolase (PARG) has endo- and exoglycosidase activities which can cleave glycosidic bonds, rapidly reversing the action of PARP enzymes. Like addition of poly(ADP-ribose) (PAR) by PARP, removal of PAR by PARG is also thought to be required for repair of DNA strand breaks and for continued replication at perturbed forks. Here we use siRNA to show a synthetic lethal relationship between PARG and BRCA1, BRCA2, PALB2, FAM175A (ABRAXAS) and BARD1. In addition, we demonstrate that MCF7 cells depleted of these proteins are sensitive to Gallotannin and a novel and specific PARG inhibitor PDD00017273. We confirm that PARG inhibition increases endogenous DNA damage, stalls replication forks and increases homologous recombination, and propose that it is the lack of homologous recombination (HR) proteins at PARG inhibitor-induced stalled replication forks that induces cell death. Interestingly not all genes that are synthetically lethal with PARP result in sensitivity to PARG inhibitors, suggesting that although there is overlap, the functions of PARP and PARG may not be completely identical. These data together add further evidence to the possibility that single treatment therapy with PARG inhibitors could be used for treatment of certain HR deficient tumours and provide insight into the relationship between PARP, PARG and the processes of DNA repair.
Collapse
Affiliation(s)
- Polly Gravells
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, United Kingdom
| | - Emma Grant
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, United Kingdom
| | - Kate M Smith
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4BX, United Kingdom
| | - Dominic I James
- Drug Discovery Unit, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4BX, United Kingdom
| | - Helen E Bryant
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, United Kingdom.
| |
Collapse
|
182
|
Zhou B, Xu JW, Cheng YG, Gao JY, Hu SY, Wang L, Zhan HX. Early detection of pancreatic cancer: Where are we now and where are we going? Int J Cancer 2017; 141:231-241. [PMID: 28240774 DOI: 10.1002/ijc.30670] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/25/2017] [Accepted: 02/20/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic cancer (PC) is one of the most lethal malignancies. Recent studies indicate that patients with incidentally diagnosed PC have better prognosis than those with symptoms and that there is a sufficient window for early detection. However, effective early diagnosis remains difficult and depends mainly on imaging modalities and the development of screening methodologies with highly sensitive and specific biomarkers. This review summarizes recent advances in effective screening for early diagnosis of PC using imaging modalities and novel molecular biomarkers discovered from various "omics" studies including genomics, epigenomics, non-coding RNA, metabonomics, liquid biopsy (CTC, ctDNA and exosomes) and microbiomes, and their use in body fluids (feces, urine and saliva). Although many biomarkers for early detection of PC have been discovered through various methods, larger scale and rigorous validation is required before their application in the clinic. In addition, more effective and specific biomarkers of PC are urgently needed.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Hepatopancreatobiliary Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, 266003, China
| | - Jian-Wei Xu
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Yu-Gang Cheng
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Jing-Yue Gao
- Department of Basic Medicine, Medical College of Shandong University, Jinan, 250012, China
| | - San-Yuan Hu
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Lei Wang
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong Province, 250012, China
| | - Han-Xiang Zhan
- Department of General Surgery, Qilu hospital, Shandong University, Jinan, Shandong Province, 250012, China
| |
Collapse
|
183
|
Manfredini M, Pellacani G, Losi L, Maccaferri M, Tomasi A, Ponti G. Desmoplastic melanoma: a challenge for the oncologist. Future Oncol 2017; 13:337-345. [DOI: 10.2217/fon-2016-0334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: To evaluate clinical, pathologic and genetic features of desmoplastic melanoma (DM). Materials & methods: Analysis of all DM records from 1991 to 2015. Results: The most common location of DMs was the head and neck (69%); median age and follow-up were 60.5 and 7.3 years, respectively. A familial predisposition for DMs and others malignancies was analyzed. Thin Breslow thickness (<4.5 mm) was associated with an intraepidermal component or a previous lentigo maligna, whereas high Breslow thickness (>4.5 mm) was observed in ‘pure’ DM. Conclusion: DM could progress from an early phase, characterized by an intraepidermal component, to late phase, characterized by a dermal nodule. This hypothesis correlates with melanoma genetic and NF1 mutation, which could be an early event in the progression of DM.
Collapse
Affiliation(s)
- Marco Manfredini
- Department of Surgical, Medical, Dental & Morphological Sciences with Interest Transplant, Oncological & Regenerative Medicine, Dermatology Unit, University of Modena & Reggio Emilia, Modena, Italy
| | - Giovanni Pellacani
- Department of Surgical, Medical, Dental & Morphological Sciences with Interest Transplant, Oncological & Regenerative Medicine, Dermatology Unit, University of Modena & Reggio Emilia, Modena, Italy
| | - Lorena Losi
- Department of Pathology, University of Modena & Reggio Emilia, Modena, Italy
| | - Monia Maccaferri
- Department of Diagnostic & Clinical Medicine & Public Health, Clinical Pathology Unit, University of Modena & Reggio Emilia, Modena, Italy
| | - Aldo Tomasi
- Department of Diagnostic & Clinical Medicine & Public Health, Clinical Pathology Unit, University of Modena & Reggio Emilia, Modena, Italy
| | - Giovanni Ponti
- Department of Diagnostic & Clinical Medicine & Public Health, Clinical Pathology Unit, University of Modena & Reggio Emilia, Modena, Italy
| |
Collapse
|
184
|
Yang XR, Rotunno M, Xiao Y, Ingvar C, Helgadottir H, Pastorino L, van Doorn R, Bennett H, Graham C, Sampson JN, Malasky M, Vogt A, Zhu B, Bianchi-Scarra G, Bruno W, Queirolo P, Fornarini G, Hansson J, Tuominen R, Burdett L, Hicks B, Hutchinson A, Jones K, Yeager M, Chanock SJ, Landi MT, Höiom V, Olsson H, Gruis N, Ghiorzo P, Tucker MA, Goldstein AM. Multiple rare variants in high-risk pancreatic cancer-related genes may increase risk for pancreatic cancer in a subset of patients with and without germline CDKN2A mutations. Hum Genet 2016; 135:1241-1249. [PMID: 27449771 PMCID: PMC5152573 DOI: 10.1007/s00439-016-1715-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/16/2016] [Indexed: 12/29/2022]
Abstract
The risk of pancreatic cancer (PC) is increased in melanoma-prone families but the causal relationship between germline CDKN2A mutations and PC risk is uncertain, suggesting the existence of non-CDKN2A factors. One genetic possibility involves patients having mutations in multiple high-risk PC-related genes; however, no systematic examination has yet been conducted. We used next-generation sequencing data to examine 24 putative PC-related genes in 43 PC patients with and 23 PC patients without germline CDKN2A mutations and 1001 controls. For each gene and the four pathways in which they occurred, we tested whether PC patients (overall or CDKN2A+ and CDKN2A- cases separately) had an increased number of rare nonsynonymous variants. Overall, we identified 35 missense variants in PC patients, 14 in CDKN2A+ and 21 in CDKN2A- PC cases. We found nominally significant associations for mismatch repair genes (MLH1, MSH2, MSH6, PMS2) in all PC patients and for ATM, CPA1, and PMS2 in CDKN2A- PC patients. Further, nine CDKN2A+ and four CDKN2A- PC patients had rare potentially deleterious variants in multiple PC-related genes. Loss-of-function variants were only observed in CDKN2A- PC patients, with ATM having the most pathogenic variants. Also, ATM variants (n = 5) were only observed in CDKN2A- PC patients with a family history that included digestive system tumors. Our results suggest that a subset of PC patients may have increased risk because of germline mutations in multiple PC-related genes.
Collapse
Affiliation(s)
- Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Melissa Rotunno
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Division of Cancer Control and Population Studies, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yanzi Xiao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Hildur Helgadottir
- Department of Oncology Pathology, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Genetics of Rare Cancers, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Remco van Doorn
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hunter Bennett
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cole Graham
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael Malasky
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Aurelie Vogt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Giovanna Bianchi-Scarra
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Genetics of Rare Cancers, IRCCS AOU San Martino-IST, Genoa, Italy
| | - William Bruno
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Genetics of Rare Cancers, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Paola Queirolo
- Medical Oncology Unit, IRCCS AOU San Martino-IST, Genoa, Italy
| | | | - Johan Hansson
- Department of Oncology Pathology, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Rainer Tuominen
- Department of Oncology Pathology, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Laurie Burdett
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Veronica Höiom
- Department of Oncology Pathology, Karolinska Institutet and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Håkan Olsson
- Department of Oncology, Lund University Hospital, Lund, Sweden
| | - Nelleke Gruis
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
- Genetics of Rare Cancers, IRCCS AOU San Martino-IST, Genoa, Italy
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
- , 9609 Medical Center Dr, Bethesda, MD, 20892-9769, USA.
| |
Collapse
|
185
|
Teo MY, O’Reilly EM. Is it time to split strategies to treat homologous recombinant deficiency in pancreas cancer? J Gastrointest Oncol 2016; 7:738-749. [PMID: 27747088 PMCID: PMC5056255 DOI: 10.21037/jgo.2016.05.04] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 04/13/2016] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is a highly lethal malignancy which tends to present with late stage disease. To date, identification of oncogenic drivers and aberrations has not led to effective targeted therapy. Approximately 5-15% of pancreatic cancer has an inheritable component. In fact, pancreatic adenocarcinoma is now recognized as a BRCA1/2-related cancer. Germline BRCA1/2 mutations can be found in up to 3.6-7% of unselected pancreatic cancer patients although the rates are significantly higher amongst patients with Ashkenazi Jewish ancestry. Germline mutations of other components of DNA repair and homologous recombination have also been identified although at much lower frequency. Large sequencing efforts have further identified somatic mutations in these genes in a small subset of pancreatic cancers. Small series and case reports have suggested that pancreatic cancers harboring BRCA1/2 or other homologous repair gene mutations demonstrate enhanced response to platinum-based chemotherapy although this has not been prospectively validated. Clinical trials with poly (ADP-ribose) polymerase (PARP) inhibitors as monotherapy or in combination with chemotherapy in different clinical settings are currently on-going. A subtype of pancreatic adenocarcinoma as characterized by deficiency in homologous recombination exists although the optimal management strategy remains to be fully elucidated.
Collapse
Affiliation(s)
| | - Eileen M. O’Reilly
- Memorial Sloan Kettering Cancer Center, NY, USA
- Weill Cornell Medical College, NY, USA
| |
Collapse
|
186
|
Abstract
Familial pancreatic cancer (FPC) includes those kindreds that contain at least two first-degree relatives with pancreatic ductal adenocarcinoma. At least 12 known hereditary syndromes or genes are associated with increased risk of developing pancreatic cancer, the foremost being BRCA2 and CDKN2A. Research into the identification of mutations in known cancer predisposition genes and through next-generation sequencing has revealed extensive heterogeneity. The development of genetic panel testing has enabled genetic risk assessment and predisposition testing to be routinely offered. Precision oncology has opened the possibility of "incidental" germline mutations that may have implications for family members. However, in both cases, evidence-based recommendations for managing patients and at-risk family members in light of genetic status remain emergent, with current practice based on expert opinion.
Collapse
Affiliation(s)
- Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic Cancer Center, Rochester, MN.
| |
Collapse
|
187
|
de Mestier L, Danset JB, Neuzillet C, Rebours V, Cros J, Soufir N, Hammel P. Pancreatic ductal adenocarcinoma in BRCA2 mutation carriers. Endocr Relat Cancer 2016; 23:T57-67. [PMID: 27511924 DOI: 10.1530/erc-16-0269] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 08/10/2016] [Indexed: 12/13/2022]
Abstract
Germline BRCA2 mutations are the first known cause of inherited (familial) pancreatic ductal adenocarcinoma (PDAC). This tumor is the third most frequent cancer in carriers of germline BRCA2 mutations, as it occurs in around 10% of BRCA2 families. PDAC is known as one of the most highly lethal cancers, mainly because of its chemoresistance and frequently late diagnosis. Based on recent developments in molecular biology, a subgroup of BRCA2-associated PDAC has been created, allowing screening, early surgical treatment and personalized systemic treatment. BRCA2 germline mutation carriers who have ≥1 first-degree relative, or ≥2 blood relatives with PDAC, should undergo screening and regular follow-up based on magnetic resonance imaging and endoscopic ultrasound. The goal of screening is to detect early invasive PDAC and advanced precancerous lesions suitable for a stepwise surgical complete (R0) resection. Increasing evidence on the molecular role of the BRCA2 protein in the homologous recombination of DNA damages suggest that BRCA2-related PDAC are sensitive to agents causing DNA cross-linking damage, such as platinum salts, and treatments targeting rescue DNA repair pathways, such as poly(ADP-ribose) polymerase inhibitors that are currently under investigation.
Collapse
Affiliation(s)
- Louis de Mestier
- Department of Gastroenterology and PancreatologyBeaujon Hospital, Paris 7 University, APHP, Clichy, France
| | - Jean-Baptiste Danset
- Department of Hepato-GastroenterologyEuropean Georges-Pompidou Hospital, APHP, Paris, France
| | - Cindy Neuzillet
- Department of Digestive OncologyBeaujon Hospital, Paris 7 University, APHP, Clichy, France
| | - Vinciane Rebours
- Department of Gastroenterology and PancreatologyBeaujon Hospital, Paris 7 University, APHP, Clichy, France
| | - Jérôme Cros
- Department of PathologyBeaujon Hospital, Paris 7 University, APHP, Clichy, France
| | - Nadem Soufir
- Department of GeneticsBichat Hospital, Paris 7 University, APHP, Clichy, France
| | - Pascal Hammel
- Department of Digestive OncologyBeaujon Hospital, Paris 7 University, APHP, Clichy, France
| |
Collapse
|
188
|
Abstract
Malignant melanoma is a rare, often fatal form of skin cancer with a complex multigenic etiology. The incidence of melanoma is increasing at an alarming rate. A number of heritable factors contribute to a patient's overall melanoma risk, including response to ultraviolet light, nevus number, and pigmentation characteristics, such as eye and hair color. Approximately 5%-10% of melanoma cases are familial, yet the majority of familial cases lack identifiable germ-line mutations in known susceptibility genes. Additionally, most familial melanomas lack germ-line mutations in genes that are commonly mutated in sporadic melanoma. Candidate and systematic genome-wide association studies have led to an improved understanding of the risk factors for melanoma and the identification of susceptibility genes. In this review, we provide an overview of the major risk factors and known genes implicated in familial melanoma susceptibility.
Collapse
Affiliation(s)
- Jason E Hawkes
- Department of Dermatology, University of Utah, Salt Lake City, UT
| | - Amanda Truong
- Department of Dermatology, University of Utah, Salt Lake City, UT
| | - Laurence J Meyer
- Department of Dermatology, University of Utah, Salt Lake City, UT; Veterans Administration Hospital, Salt Lake City, UT.
| |
Collapse
|
189
|
Tung N, Domchek SM, Stadler Z, Nathanson KL, Couch F, Garber JE, Offit K, Robson ME. Counselling framework for moderate-penetrance cancer-susceptibility mutations. Nat Rev Clin Oncol 2016; 13:581-8. [PMID: 27296296 PMCID: PMC5513673 DOI: 10.1038/nrclinonc.2016.90] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The use of multigene panels for the assessment of cancer susceptibility is expanding rapidly in clinical practice, particularly in the USA, despite concerns regarding the uncertain clinical validity for some gene variants and the uncertain clinical utility of most multigene panels. So-called 'moderate-penetrance' gene mutations associated with cancer susceptibility are identified in approximately 2-5% of individuals referred for clinical testing; some of these mutations are potentially actionable. Nevertheless, the appropriate management of individuals harbouring such moderate-penetrance genetic variants is unclear. The cancer risks associated with mutations in moderate-penetrance genes are lower and different than those reported for high-penetrance gene mutations (such as mutations in BRCA1 and BRCA2, and those associated with Lynch syndrome). The extrapolation of guidelines for the management of individuals with high-penetrance variants of cancer-susceptibility genes to the clinical care of patients with moderate-penetrance gene mutations could result in substantial harm. Thus, we provide a framework for clinical decision-making pending the development of a sufficient evidence base to document the clinical utility of the interventions for individuals with inherited moderate-penetrance gene mutations associated with an increased risk of cancer.
Collapse
Affiliation(s)
- Nadine Tung
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Susan M Domchek
- Abramson Cancer Center, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, USA, and the Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Zsofia Stadler
- Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue; and the Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| | - Katherine L Nathanson
- Abramson Cancer Center, 3400 Spruce Street, Philadelphia, Pennsylvania 19104, USA, and the Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA
| | - Fergus Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street South West, Rochester, Minnesota 55905, USA
| | - Judy E Garber
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Kenneth Offit
- Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue; and the Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| | - Mark E Robson
- Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, 1275 York Avenue; and the Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| |
Collapse
|
190
|
Abstract
Although almost all gastrointestinal cancers develop from sporadic genomic events, approximately 5% arise from germline mutations in genes associated with cancer predisposition. The number of these genes continues to increase. Tumor phenotypes and family history provide the framework for identifying at-risk individuals. The diagnosis of a hereditary cancer syndrome has implications for management of patients and their families. Systematic approaches that integrate family history and molecular characterization of tumors and polyps facilitate identification of individuals with this genetic predisposition. This article summarizes diagnosis and management of hereditary cancer syndromes associated with gastrointestinal cancers.
Collapse
Affiliation(s)
- Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Health System, 2150A Cancer Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
191
|
Mogilyansky E, Clark P, Quann K, Zhou H, Londin E, Jing Y, Rigoutsos I. Post-transcriptional Regulation of BRCA2 through Interactions with miR-19a and miR-19b. Front Genet 2016; 7:143. [PMID: 27630665 PMCID: PMC5005319 DOI: 10.3389/fgene.2016.00143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 07/21/2016] [Indexed: 12/31/2022] Open
Abstract
Breast cancer type 2, early onset susceptibility gene (BRCA2) is a major component of the homologous recombination DNA repair pathway. It acts as a tumor suppressor whose function is often lost in cancers. Patients with specific mutations in the BRCA2 gene often display discrete clinical, histopathological, and molecular features. However, a subset of sporadic cancers has wild type BRCA2 and display defects in the homology-directed repair pathway, which is the hallmark of ‘BRCAness.’ The mechanisms by which BRCAness arises are not well understood but post-transcriptional regulation of BRCA2 gene expression by microRNAs (miRNAs) may contribute to this phenotype. Here, we examine the post-transcriptional effects that some members of the six-miRNA cluster known as the miR-17/92 cluster have on the abundance of BRCA2’s messenger RNA (mRNA) and protein. We discuss two interactions involving the miR-19a and miR-19b members of the cluster and the 3′UTR of BRCA2’s mRNA. We investigated these miRNA:mRNA interactions in 15 cell lines derived from pancreatic, breast, colon, and kidney tissue. We show that over-expression of these two miRNAs results in a concomitant decrease of BRCA2’s mRNA and protein expression in a subset of the tested cell lines. Additionally, using luciferase reporter assays we identified direct interactions between miR-19a/miR-19b and a miRNA response element (MRE) in BRCA2’s 3′UTR. Our results suggest that BRCA2 is subject to a complex post-transcriptional regulatory program that has specific dependencies on the genetic and phenotypic background of cell types.
Collapse
Affiliation(s)
- Elena Mogilyansky
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Peter Clark
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia PA, USA
| | - Kevin Quann
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Honglei Zhou
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Eric Londin
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Yi Jing
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| | - Isidore Rigoutsos
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia PA, USA
| |
Collapse
|
192
|
Murata S, Zhang C, Finch N, Zhang K, Campo L, Breuer EK. Predictors and Modulators of Synthetic Lethality: An Update on PARP Inhibitors and Personalized Medicine. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2346585. [PMID: 27642590 PMCID: PMC5013223 DOI: 10.1155/2016/2346585] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors have proven to be successful agents in inducing synthetic lethality in several malignancies. Several PARP inhibitors have reached clinical trial testing for treatment in different cancers, and, recently, Olaparib (AZD2281) has gained both United States Food and Drug Administration (USFDA) and the European Commission (EC) approval for use in BRCA-mutated advanced ovarian cancer treatment. The need to identify biomarkers, their interactions in DNA damage repair pathways, and their potential utility in identifying patients who are candidates for PARP inhibitor treatment is well recognized. In this review, we detail many of the biomarkers that have been investigated for their ability to predict both PARP inhibitor sensitivity and resistance in preclinical studies as well as the results of several clinical trials that have tested the safety and efficacy of different PARP inhibitor agents in BRCA and non-BRCA-mutated cancers.
Collapse
Affiliation(s)
- Stephen Murata
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Catherine Zhang
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Nathan Finch
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Kevin Zhang
- Department of Otorhinolaryngology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Loredana Campo
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Eun-Kyoung Breuer
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
193
|
Peters MLB, Tseng JF, Miksad RA. Genetic Testing in Pancreatic Ductal Adenocarcinoma: Implications for Prevention and Treatment. Clin Ther 2016; 38:1622-35. [DOI: 10.1016/j.clinthera.2016.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/20/2022]
|
194
|
Hackeng WM, Hruban RH, Offerhaus GJA, Brosens LAA. Surgical and molecular pathology of pancreatic neoplasms. Diagn Pathol 2016; 11:47. [PMID: 27267993 PMCID: PMC4897815 DOI: 10.1186/s13000-016-0497-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/28/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Histologic characteristics have proven to be very useful for classifying different types of tumors of the pancreas. As a result, the major tumor types in the pancreas have long been classified based on their microscopic appearance. MAIN BODY Recent advances in whole exome sequencing, gene expression profiling, and knowledge of tumorigenic pathways have deepened our understanding of the underlying biology of pancreatic neoplasia. These advances have not only confirmed the traditional histologic classification system, but also opened new doors to early diagnosis and targeted treatment. CONCLUSION This review discusses the histopathology, genetic and epigenetic alterations and potential treatment targets of the five major malignant pancreatic tumors - pancreatic ductal adenocarcinoma, pancreatic neuroendocrine tumor, solid-pseudopapillary neoplasm, acinar cell carcinoma and pancreatoblastoma.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Carcinoma, Acinar Cell/diagnosis
- Carcinoma, Acinar Cell/genetics
- Carcinoma, Acinar Cell/surgery
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/surgery
- Eye Diseases, Hereditary/diagnosis
- Eye Diseases, Hereditary/genetics
- Eye Diseases, Hereditary/surgery
- Humans
- Neuroendocrine Tumors/diagnosis
- Neuroendocrine Tumors/genetics
- Neuroendocrine Tumors/surgery
- Optic Nerve Diseases/diagnosis
- Optic Nerve Diseases/genetics
- Optic Nerve Diseases/surgery
- Pancreas/pathology
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/surgery
Collapse
Affiliation(s)
- Wenzel M Hackeng
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G Johan A Offerhaus
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Lodewijk A A Brosens
- Department of Pathology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
195
|
Borecka M, Zemankova P, Vocka M, Soucek P, Soukupova J, Kleiblova P, Sevcik J, Kleibl Z, Janatova M. Mutation analysis of the PALB2 gene in unselected pancreatic cancer patients in the Czech Republic. Cancer Genet 2016; 209:199-204. [DOI: 10.1016/j.cancergen.2016.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/10/2016] [Accepted: 03/21/2016] [Indexed: 12/19/2022]
|
196
|
|
197
|
Joergensen MT, Gerdes AM, Sorensen J, Schaffalitzky de Muckadell O, Mortensen MB. Is screening for pancreatic cancer in high-risk groups cost-effective? - Experience from a Danish national screening program. Pancreatology 2016; 16:584-92. [PMID: 27090585 DOI: 10.1016/j.pan.2016.03.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pancreatic cancer (PC) is the fourth leading cause of cancer death worldwide, symptoms are few and diffuse, and when the diagnosis has been made only 10-15% would benefit from resection. Surgery is the only potentially curable treatment for pancreatic cancer, and the prognosis seems to improve with early detection. A hereditary component has been identified in 1-10% of the PC cases. To comply with this, screening for PC in high-risk groups with a genetic disposition for PC has been recommended in research settings. DESIGN Between January 2006 and February 2014 31 patients with Hereditary pancreatitis or with a disposition of HP and 40 first-degree relatives of patients with Familial Pancreatic Cancer (FPC) were screened for development of Pancreatic Ductal Adenocarcinoma (PDAC) with yearly endoscopic ultrasound. The cost-effectiveness of screening in comparison with no-screening was assessed by the incremental cost-utility ratio (ICER). RESULTS By screening the FPC group we identified 2 patients with PDAC who were treated by total pancreatectomy. One patient is still alive, while the other died after 7 months due to cardiac surgery complications. Stratified analysis of patients with HP and FPC provided ICERs of 47,156 US$ vs. 35,493 US$ per life-year and 58,647 US$ vs. 47,867 US$ per QALY. Including only PDAC related death changed the ICER to 31,722 US$ per life-year and 42,128 US$ per QALY. The ICER for patients with FPC was estimated at 28,834 US$ per life-year and 38,785 US$ per QALY. CONCLUSIONS With a threshold value of 50,000 US$ per QALY this screening program appears to constitute a cost-effective intervention although screening of HP patients appears to be less cost-effective than FPC patients.
Collapse
Affiliation(s)
- Maiken Thyregod Joergensen
- Vejle Hospital, Southern Denmark, Odense, Denmark; Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.
| | | | - Jan Sorensen
- Centre for Health Economic Research (COHERE), Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
198
|
Caini S, Radice D, Tosti G, Spadola G, Cocorocchio E, Ferrucci PF, Testori A, Pennacchioli E, Fargnoli MC, Palli D, Bazolli B, Botteri E, Gandini S. Risk of second primary malignancies among 1537 melanoma patients and risk of second primary melanoma among 52 354 cancer patients in Northern Italy. J Eur Acad Dermatol Venereol 2016; 30:1491-6. [PMID: 26990937 DOI: 10.1111/jdv.13645] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/07/2016] [Accepted: 02/15/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND The number of melanoma survivors has been increasing for decades due to early diagnosis and improved survival. These patients have an increased risk of developing a second primary cancer (SPC); also, melanoma is frequently diagnosed among patients firstly diagnosed with an extracutaneous malignancy. OBJECTIVE We evaluated the risk of developing a SPC among 1537 melanoma patients, and the risk of second primary melanoma (SPM) in 52 354 extracutaneous cancer patients, who were treated at the European Institute of Oncology in Milan, Italy, during 2000-2010. MATERIAL AND METHODS We calculated standardized incidence ratios (SIR) by applying gender-, age-, year- and region-specific reference rates to the follow-up time accrued between the diagnosis of the first and the second primary malignancies. RESULTS Seventy-six SPC were diagnosed during a median follow-up of 4 years, of which 49 (64%) during the first 2 years upon melanoma diagnosis. The SIR was increased for cancer of breast (4.10, 95% CI 2.79-6.03), thyroid (4.67, 95% CI 1.94-11.22), brain (6.13, 95% CI 2.30-16.33) and for non-Hodgkin lymphoma (3.12, 95% CI 1.30-7.50). During a median follow-up of 4 years, 127 SPM were diagnosed: thick lesions were less frequent than for melanoma diagnosed as first cancer. The SIR was increased for cancer of breast (5.13, 95%CI 3.91-6.73), thyroid (16.2, 95%CI: 5.22-50.2), head and neck (5.62, 95%CI 1.41-22.50), soft tissue (8.68, 95%CI 2.17-34.70), cervix (12.5, 95% CI 3.14-50.20), kidney (3.19, 95%CI 1.52-6.68), prostate (4.36, 95%CI 2.63-7.24) and acute myeloid leukaemia (6.44, 95%CI 2.42-17.20). CONCLUSIONS The most likely causes of these associations are the clustering of lifestyle risk factors in the same subgroups of population, mainly on a sociocultural basis and surveillance bias. This raises important questions about how to best follow cancer survivors by avoiding an inefficient use of resources and an excessive medicalization of these patients' lives.
Collapse
Affiliation(s)
- S Caini
- Unit of Cancer Risk Factors and Lifestyle Epidemiology, Institute for Cancer Research and Prevention, Florence, Italy
| | - D Radice
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - G Tosti
- Division of Dermatoncological Surgery, European Institute of Oncology, Milan, Italy
| | - G Spadola
- Division of Dermatoncological Surgery, European Institute of Oncology, Milan, Italy
| | - E Cocorocchio
- Medical Oncology of Melanoma Unit, European Institute of Oncology, Milan, Italy
| | - P F Ferrucci
- Medical Oncology of Melanoma Unit, European Institute of Oncology, Milan, Italy
| | - A Testori
- Division of Dermatoncological Surgery, European Institute of Oncology, Milan, Italy
| | - E Pennacchioli
- Division of Melanoma and Muscle-Cutaneous Sarcomas, European Institute of Oncology, Milan, Italy
| | - M C Fargnoli
- Department of Dermatology, University of L'Aquila, L'Aquila, Italy
| | - D Palli
- Unit of Cancer Risk Factors and Lifestyle Epidemiology, Institute for Cancer Research and Prevention, Florence, Italy
| | - B Bazolli
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - E Botteri
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - S Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| |
Collapse
|
199
|
Morphological heterogeneity in ductal adenocarcinoma of the pancreas - Does it matter? Pancreatology 2016; 16:295-301. [PMID: 26924665 DOI: 10.1016/j.pan.2016.02.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/07/2016] [Accepted: 02/08/2016] [Indexed: 02/08/2023]
Abstract
Morphological heterogeneity is a common finding in pancreatic ductal adenocarcinoma. Inter- and intra-tumour heterogeneity relates not only to the microscopic appearances of the tumour cell population, but pertains also to other essential aspects of the cancer, including the grade of differentiation, growth pattern and desmoplastic stroma. While the existence of considerable morphological variation is well known among pathologists, it is usually not fully appreciated by the wider community. Morphological heterogeneity in pancreatic cancer is only partially represented in the WHO classification, and current pathology guidelines do not recommend reporting on morphological variation other than the conventional variants of ductal adenocarcinoma. Although tumour heterogeneity is increasingly recognized as a major determinant of therapeutic response, morphological heterogeneity has been left unconsidered as a possible proxy for underlying aberrations - genomic or otherwise - that determine the effect of treatment. Various aspects of morphological heterogeneity in pancreatic ductal adenocarcinoma are illustrated in this article and discussed along with the possible implications for patient management and research.
Collapse
|
200
|
Abstract
Although relatively rare, pancreatic tumors are highly lethal [1]. In the United States, an estimated 48,960 individuals will be diagnosed with pancreatic cancer and 40,560 will die from this disease in 2015 [1]. Globally, 337,872 new pancreatic cancer cases and 330,391 deaths were estimated in 2012 [2]. In contrast to most other cancers, mortality rates for pancreatic cancer are not improving; in the US, it is predicted to become the second leading cause of cancer related deaths by 2030 [3, 4]. The vast majority of tumors arise in the exocrine pancreas, with pancreatic ductal adenocarcinoma (PDAC) accounting for approximately 95% of tumors. Tumors arising in the endocrine pancreas (pancreatic neuroendocrine tumors) represent less than 5% of all pancreatic tumors [5]. Smoking, type 2 diabetes mellitus (T2D), obesity and pancreatitis are the most consistent epidemiological risk factors for pancreatic cancer [5]. Family history is also a risk factor for developing pancreatic cancer with odds ratios (OR) ranging from 1.7-2.3 for first-degree relatives in most studies, indicating that shared genetic factors may play a role in the etiology of this disease [6-9]. This review summarizes the current knowledge of germline pancreatic cancer risk variants with a special emphasis on common susceptibility alleles identified through Genome Wide Association Studies (GWAS).
Collapse
Affiliation(s)
- Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|