1
|
Archasappawat S, Al-Musawi F, Liu P, Lee E, Hwang CI. Familial Pancreatic Cancer Research: Bridging Gaps in Basic Research and Clinical Application. Biomolecules 2024; 14:1381. [PMID: 39595558 PMCID: PMC11592027 DOI: 10.3390/biom14111381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Familial pancreatic cancer (FPC) represents a significant yet underexplored area in pancreatic cancer research. Basic research efforts are notably limited, and when present, they are predominantly centered on the BRCA1 and BRCA2 mutations due to the scarcity of other genetic variants associated with FPC, leading to a limited understanding of the broader genetic landscape of FPC. This review examines the current state of FPC research, focusing on the molecular mechanisms driving pancreatic ductal adenocarcinoma (PDAC) progression. It highlights the role of homologous recombination (HR) and its therapeutic exploitation via synthetic lethality with PARP inhibitors in BRCA1/2-deficient tumors. The review discusses various pre-clinical models of FPC, including conventional two-dimensional (2D) cell lines, patient-derived organoids (PDOs), patient-derived xenografts (PDXs), and genetically engineered mouse models (GEMMs), as well as new advancements in FPC research.
Collapse
Affiliation(s)
- Suyakarn Archasappawat
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Fatimah Al-Musawi
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Peiyi Liu
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - EunJung Lee
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Chang-il Hwang
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
2
|
White MJ, Sheka AC, LaRocca CJ, Irey RL, Ma S, Wirth KM, Benner A, Denbo JW, Jensen EH, Ankeny JS, Ikramuddin S, Tuttle TM, Hui JYC, Marmor S. The association of new-onset diabetes with subsequent diagnosis of pancreatic cancer-novel use of a large administrative database. J Public Health (Oxf) 2023; 45:e266-e274. [PMID: 36321614 PMCID: PMC10273390 DOI: 10.1093/pubmed/fdac118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Screening options for pancreatic ductal adenocarcinoma (PDAC) are limited. New-onset type 2 diabetes (NoD) is associated with subsequent diagnosis of PDAC in observational studies and may afford an opportunity for PDAC screening. We evaluated this association using a large administrative database. METHODS Patients were identified using claims data from the OptumLabs® Data Warehouse. Adult patients with NoD diagnosis were matched 1:3 with patients without NoD using age, sex and chronic obstructive pulmonary disease (COPD) status. The event of PDAC diagnosis was compared between cohorts using the Kaplan-Meier method. Factors associated with PDAC diagnosis were evaluated with Cox's proportional hazards modeling. RESULTS We identified 640 421 patients with NoD and included 1 921 263 controls. At 3 years, significantly more PDAC events were identified in the NoD group vs control group (579 vs 505; P < 0.001). When controlling for patient factors, NoD was significantly associated with elevated risk of PDAC (HR 3.474, 95% CI 3.082-3.920, P < 0.001). Other factors significantly associated with PDAC diagnosis were increasing age, increasing age among Black patients, and COPD diagnosis (P ≤ 0.05). CONCLUSIONS NoD was independently associated with subsequent diagnosis of PDAC within 3 years. Future studies should evaluate the feasibility and benefit of PDAC screening in patients with NoD.
Collapse
Affiliation(s)
- M J White
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
| | - A C Sheka
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- OptumLabs® Visiting Fellow, Eden Prairie, MN, USA Institute for Health Informatics, University of Minnesota, Minneapolis MN, 55455 USA
| | - C J LaRocca
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - R L Irey
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| | - S Ma
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| | - K M Wirth
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- OptumLabs® Visiting Fellow, Eden Prairie, MN, USA Institute for Health Informatics, University of Minnesota, Minneapolis MN, 55455 USA
| | - A Benner
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| | - J W Denbo
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa FL 33612 USA
| | - E H Jensen
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - J S Ankeny
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - S Ikramuddin
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- OptumLabs® Visiting Fellow, Eden Prairie, MN, USA Institute for Health Informatics, University of Minnesota, Minneapolis MN, 55455 USA
| | - T M Tuttle
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - J Y C Hui
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
| | - S Marmor
- Department of Surgery, University of Minnesota, Minneapolis MN, 55455 USA
- Masonic Cancer Center, University of Minnesota, Minneapolis MN 55455, USA
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis MN, 55455 USA
| |
Collapse
|
3
|
A Genome-First Approach to Estimate Prevalence of Germline Pathogenic Variants and Risk of Pancreatic Cancer in Select Cancer Susceptibility Genes. Cancers (Basel) 2022; 14:cancers14133257. [PMID: 35805029 PMCID: PMC9265005 DOI: 10.3390/cancers14133257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 12/20/2022] Open
Abstract
Patients with germline pathogenic variants (GPV) in cancer predisposition genes are at increased risk of pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer. The genes most frequently found to harbor GPV in unselected PDAC cases are ATM, BRCA1, BRCA2, CDKN2A, CHEK2, and PALB2. However, GPV prevalence and gene-specific associations have not been extensively studied in the general population. To further explore these associations, we analyzed genomic and phenotypic data obtained from the UK Biobank (UKB) and Geisinger MyCode Community Health Initiative (GHS) cohorts comprising 200,600 and 175,449 participants, respectively. We estimated the frequency and calculated relative risks (RRs) of heterozygotes in both cohorts and a subset of individuals with PDAC. The combined frequency of heterozygous carriers of GPV in the general population ranged from 1.22% for CHEK2 to 0.05% for CDKN2A. The frequency of GPV in PDAC cases varied from 2.38% (ATM) to 0.19% (BRCA1 and CDKN2A). The RRs of PDAC were elevated for all genes except for BRCA1 and varied widely by gene from high (ATM) to low (CHEK2, BRCA2). This work expands our understanding of the frequencies of GPV heterozygous carriers and associations between PDAC and GPV in several important PDAC susceptibility genes.
Collapse
|
4
|
Kim M, Kim H, Han Y, Sohn H, Lee M, Kang YH, Kim HS, Kwon W, Jang JY. The incidence and clinical features of familial pancreatic cancer in Korea. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2022; 29:659-669. [PMID: 35064645 DOI: 10.1002/jhbp.1117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND A history of familial pancreatic cancer (FPC) increases the incidence of pancreatic cancer (PC) among first-degree relatives. We aimed to determine the incidence of FPC and analyze its clinical characteristics. METHODS Between 2010 and 2014, 1159 patients with PC were included in the study. We evaluated the incidence of FPC, clinicopathological features, and survival prognosis between FPC and non-FPC patients. We further analyzed the clinical outcomes of 389 patients with PC who underwent curative-intent surgery. RESULTS Familial pancreatic cancer incidence was 3.1% (n = 36) among all patients with PC (n = 1159). FPC was diagnosed at an advanced clinical stage compared to non-FPC (P = .041). The tested variables and 5-year survival rate (5YSR) between FPC and non-FPC after propensity score matching had no differences (5YSR: 4.6% vs 2.6%, P = .834). Among PC patients who underwent curative-intent surgery (n = 389), FPC incidence was 1.8% (n = 7). FPC patients were older than non-FPC patients (75.3 ± 4.7 years vs 64.0 ± 9.9 years, P < .001). 5YSR tended to differ between FPC and non-FPC (14.3% vs 22.5%, P = .07) groups. CONCLUSION Familial pancreatic cancer is diagnosed at an advanced stage, and FPC that has undergone resection is associated with older age or worse prognosis. A prospective nationwide pedigree registration system was required.
Collapse
Affiliation(s)
- Minseob Kim
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hongbeom Kim
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Youngmin Han
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Heeju Sohn
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Mirang Lee
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Hyung Kang
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyeong Seok Kim
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Wooil Kwon
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Jin-Young Jang
- Departments of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
5
|
Overbeek KA, Levink IJM, Koopmann BDM, Harinck F, Konings ICAW, Ausems MGEM, Wagner A, Fockens P, van Eijck CH, Groot Koerkamp B, Busch ORC, Besselink MG, Bastiaansen BAJ, van Driel LMJW, Erler NS, Vleggaar FP, Poley JW, Cahen DL, van Hooft JE, Bruno MJ. Long-term yield of pancreatic cancer surveillance in high-risk individuals. Gut 2022; 71:1152-1160. [PMID: 33820756 PMCID: PMC9120399 DOI: 10.1136/gutjnl-2020-323611] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We aimed to determine the long-term yield of pancreatic cancer surveillance in hereditary predisposed high-risk individuals. DESIGN From 2006 to 2019, we prospectively enrolled asymptomatic individuals with an estimated 10% or greater lifetime risk of pancreatic ductal adenocarcinoma (PDAC) after obligatory evaluation by a clinical geneticist and genetic testing, and subjected them to annual surveillance with both endoscopic ultrasonography (EUS) and MRI/cholangiopancreatography (MRI/MRCP) at each visit. RESULTS 366 individuals (201 mutation-negative familial pancreatic cancer (FPC) kindreds and 165 PDAC susceptibility gene mutation carriers; mean age 54 years, SD 9.9) were followed for 63 months on average (SD 43.2). Ten individuals developed PDAC, of which four presented with a symptomatic interval carcinoma and six underwent resection. The cumulative PDAC incidence was 9.3% in the mutation carriers and 0% in the FPC kindreds (p<0.001). Median PDAC survival was 18 months (range 1-32). Surgery was performed in 17 individuals (4.6%), whose pathology revealed 6 PDACs (3 T1N0M0), 7 low-grade precursor lesions, 2 neuroendocrine tumours <2 cm, 1 autoimmune pancreatitis and in 1 individual no abnormality. There was no surgery-related mortality. EUS detected more solid lesions than MRI/MRCP (100% vs 22%, p<0.001), but less cystic lesions (42% vs 83%, p<0.001). CONCLUSION The diagnostic yield of PDAC was substantial in established high-risk mutation carriers, but non-existent in the mutation-negative proven FPC kindreds. Nevertheless, timely identification of resectable lesions proved challenging despite the concurrent use of two imaging modalities, with EUS outperforming MRI/MRCP. Overall, surveillance by imaging yields suboptimal results with a clear need for more sensitive diagnostic markers, including biomarkers.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Iris J M Levink
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Brechtje D M Koopmann
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Femme Harinck
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ingrid C A W Konings
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Margreet G E M Ausems
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Casper H van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Olivier R C Busch
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Barbara A J Bastiaansen
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lydi M J W van Driel
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicole S Erler
- Department of Biostatistics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank P Vleggaar
- Department of Gastroenterology & Hepatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan-Werner Poley
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Rabe KG, Stevens MA, Hernández AT, Chandra S, Hubbard JM, Kemppainen JL, Majumder S, Petersen GM. Pancreatic cancer risk to siblings of probands in bilineal cancer settings. Genet Med 2022; 24:1008-1016. [PMID: 35227607 PMCID: PMC9326771 DOI: 10.1016/j.gim.2022.01.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Pancreatic cancer (PC) risk is increased in families, but PC risk and risk perception have been understudied when both parents have cancer. METHODS An unbiased method defining cancer triads (proband with PC and both parents with cancer) in a prospective registry estimated risk of PC to probands' siblings in triad group 1 (no parent with PC), group 2 (1 parent with PC), and group 3 (both parents with PC). We estimated standardized incidence ratios (SIRs) using a Surveillance, Epidemiology, and End Results (SEER) reference. We also estimated the risk when triad probands carried germline pathogenic/likely pathogenic variants in any of the 6 PC-associated genes (ATM, BRCA1, BRCA2, CDKN2A, MLH1, and TP53). PC risk perception/concern was surveyed in siblings and controls. RESULTS Risk of PC was higher (SIR = 3.5; 95% CI = 2.2-5.2) in 933 at-risk siblings from 297 triads. Risk increased by triad group: 2.8 (95% CI = 1.5-4.5); 4.5 (95% CI = 1.6-9.7); and 21.2 (95% CI = 4.3-62.0). SIR in variant-negative triads was 3.0 (95% CI = 1.6-5.0), whereas SIR in variant-positive triads was 10.0 (95% CI = 3.2-23.4). Siblings' perceived risk/concern of developing PC increased by triad group. CONCLUSION Sibling risks were 2.8- to 21.2-fold higher than that of the general population. Positive variant status increased the risk in triads. Increasing number of PC cases in a triad was associated with increased concern and perceived PC risk.
Collapse
Affiliation(s)
- Kari G Rabe
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Maria A Stevens
- Division of Health Care Policy and Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Amanda Toledo Hernández
- School of Medicine, Medical Science Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Shruti Chandra
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | | | | | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Gloria M Petersen
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN.
| |
Collapse
|
7
|
[Diagnostics and clinical management of premalignant diseases of the pancreas]. Internist (Berl) 2022; 63:401-413. [PMID: 35234978 DOI: 10.1007/s00108-022-01308-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
Abstract
Pancreatic cancer is one of the most aggressive solid tumors and still has a poor prognosis. A delayed diagnosis at advanced stages and a poor response to systemic treatment frequently make a curative treatment impossible. Therefore, the identification of high-risk patients and screening them regularly is the most promising approach to improve the prognosis. Chronic pancreatitis as well as neoplastic pancreatic cysts can greatly increase the risk of developing pancreatic cancer. Furthermore, familial syndromes and germline mutations also confer an increased risk for development of pancreatic cancer. This article provides an overview of the various premalignant diseases of the pancreas. The value of the various imaging modalities, such as magnetic resonance imaging and endosonography are particularly discussed as well as the screening interval and the indications for surgical treatment are explained.
Collapse
|
8
|
Principe DR. Precision Medicine for BRCA/PALB2-Mutated Pancreatic Cancer and Emerging Strategies to Improve Therapeutic Responses to PARP Inhibition. Cancers (Basel) 2022; 14:cancers14040897. [PMID: 35205643 PMCID: PMC8869830 DOI: 10.3390/cancers14040897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary For the small subset of pancreatic ductal adenocarcinoma (PDAC) patients with loss-of-function mutations to BRCA1/2 or PALB2, both first-line and maintenance therapy differs significantly. These mutations confer a loss of double-strand break DNA homologous recombination (HR), substantially altering drug sensitivities. In this review, we discuss the current treatment guidelines for PDAC tumors deficient in HR, as well as newly emerging strategies to improve drug responses in this population. We also highlight additional patient populations in which these strategies may also be effective, and novel strategies aiming to confer similar drug sensitivity to tumors proficient in HR repair. Abstract Pancreatic cancer is projected to become the second leading cause of cancer-related death by 2030. As patients typically present with advanced disease and show poor responses to broad-spectrum chemotherapy, overall survival remains a dismal 10%. This underscores an urgent clinical need to identify new therapeutic approaches for PDAC patients. Precision medicine is now the standard of care for several difficult-to-treat cancer histologies. Such approaches involve the identification of a clinically actionable molecular feature, which is matched to an appropriate targeted therapy. Selective poly (ADP-ribose) polymerase (PARP) inhibitors such as Niraparib, Olaparib, Talazoparib, Rucaparib, and Veliparib are now approved for several cancers with loss of high-fidelity double-strand break homologous recombination (HR), namely those with deleterious mutations to BRCA1/2, PALB2, and other functionally related genes. Recent evidence suggests that the presence of such mutations in pancreatic ductal adenocarcinoma (PDAC), the most common and lethal pancreatic cancer histotype, significantly alters drug responses both with respect to first-line chemotherapy and maintenance therapy. In this review, we discuss the current treatment paradigm for PDAC tumors with confirmed deficits in double-strand break HR, as well as emerging strategies to both improve responses to PARP inhibition in HR-deficient PDAC and confer sensitivity to tumors proficient in HR repair.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
9
|
Takaji R, Yamada Y, Shimada R, Matsumoto S, Daa T, Endo Y, Inomata M, Asayama Y. Retrospective evaluation of venous phase contrast-enhanced computed tomography images in patients who developed pancreatic adenocarcinomas after treatment for nonpancreatic primary cancer. BJR Open 2021; 3:20200069. [PMID: 34381945 PMCID: PMC8320134 DOI: 10.1259/bjro.20200069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 11/27/2022] Open
Abstract
Objectives: To clarify venous phase contrast-enhanced CT findings in early pancreatic adenocarcinomas by retrospectively evaluating CT images of pancreatic adenocarcinomas that developed during follow-up after treatment for non-pancreatic cancers. Methods: The study cohort comprised six patients who developed pancreatic adenocarcinomas between April 2005 and April 2020 during follow-up after treatment for non-pancreatic primary cancers. Two radiologists retrospectively evaluated CT images and reached consensus on previously reported CT findings that were suggestive of small pancreatic adenocarcinomas; namely pancreatic duct interruption and dilatation, pancreatic parenchymal atrophy, focal hypoattenuated areas, and appearance of cystic lesions. Time intervals between the first CT with these suggestive findings and the latest pre-operative CT were recorded. Doubling times were calculated in patients with hypoattenuated areas on initial CT scans. Results: Small (<10 mm) focal hypoattenuated areas with (n = 2) or without rim enhancement (n = 1) were identified on initial CT images of three patients. Pancreatic duct interruption and dilatation, pancreatic parenchymal atrophy, and cystic lesion were identified in two, one and one patient, respectively. Time intervals between initial and latest preoperative CT examination were 6–19 months (median, 14.5 months). Tumor doubling time according to CT findings was calculated as 46–407 days (median 106 days). Conclusion: Venous phase contrast-enhanced CT can provide findings that are suggestive of early pancreatic adenocarcinoma. Pancreatic phase contrast-enhanced CT should therefore be performed in patients with such findings with the aim of early detection of pancreatic adenocarcinoma. Advances in knowledge: Pancreatic adenocarcinoma can develop subsequently in patients with non-pancreatic malignancies. Patients with non-pancreatic cancers are often followed up with monophasic contrast-enhanced CT in venous phase timing. Venous phase contrast-enhanced CT can provide some findings suggestive of early pancreatic adenocarcinoma. Knowledge of these findings is important for early detection of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ryo Takaji
- Department of Radiology, Oita University Faculty of Medicine, Oita, Japan
| | - Yasunari Yamada
- Department of Radiology, Oita Red Cross Hospital, Oita, Japan
| | - Ryuichi Shimada
- Department of Radiology, Oita University Faculty of Medicine, Oita, Japan
| | | | - Tsutomu Daa
- Department of Diagnostic Pathology, Oita University Faculty of Medicine, Oita, Japan
| | - Yuichi Endo
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Oita, Japan
| | - Masafumi Inomata
- Department of Gastroenterological and Pediatric Surgery, Oita University Faculty of Medicine, Oita, Japan
| | - Yoshiki Asayama
- Department of Radiology, Oita University Faculty of Medicine, Oita, Japan
| |
Collapse
|
10
|
Liotta L, Lange S, Maurer HC, Olive KP, Braren R, Pfarr N, Burger S, Muckenhuber A, Jesinghaus M, Steiger K, Weichert W, Friess H, Schmid R, Algül H, Jost PJ, Ramser J, Fischer C, Quante AS, Reichert M, Quante M. PALLD mutation in a European family conveys a stromal predisposition for familial pancreatic cancer. JCI Insight 2021; 6:141532. [PMID: 33764904 PMCID: PMC8119201 DOI: 10.1172/jci.insight.141532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 03/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUNDPancreatic cancer is one of the deadliest cancers, with low long-term survival rates. Despite recent advances in treatment, it is important to identify and screen high-risk individuals for cancer prevention. Familial pancreatic cancer (FPC) accounts for 4%-10% of pancreatic cancers. Several germline mutations are related to an increased risk and might offer screening and therapy options. In this study, we aimed to identity of a susceptibility gene in a family with FPC.METHODSWhole exome sequencing and PCR confirmation was performed on the surgical specimen and peripheral blood of an index patient and her sister in a family with high incidence of pancreatic cancer, to identify somatic and germline mutations associated with familial pancreatic cancer. Compartment-specific gene expression data and immunohistochemistry were also queried.RESULTSThe identical germline mutation of the PALLD gene (NM_001166108.1:c.G154A:p.D52N) was detected in the index patient with pancreatic cancer and the tumor tissue of her sister. Whole genome sequencing showed similar somatic mutation patterns between the 2 sisters. Apart from the PALLD mutation, commonly mutated genes that characterize pancreatic ductal adenocarcinoma were found in both tumor samples. However, the 2 patients harbored different somatic KRAS mutations (G12D and G12V). Healthy siblings did not have the PALLD mutation, indicating a disease-specific impact. Compartment-specific gene expression data and IHC showed expression in cancer-associated fibroblasts (CAFs).CONCLUSIONWe identified a germline mutation of the palladin (PALLD) gene in 2 siblings in Europe, affected by familial pancreatic cancer, with a significant overexpression in CAFs, suggesting that stromal palladin could play a role in the development, maintenance, and/or progression of pancreatic cancer.FUNDINGDFG SFB 1321.
Collapse
Affiliation(s)
- Lucia Liotta
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Sebastian Lange
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - H. Carlo Maurer
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Kenneth P. Olive
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Rickmer Braren
- Institut für diagnostische und interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Nicole Pfarr
- Institut für Pathologie und pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Sebastian Burger
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alexander Muckenhuber
- Institut für Pathologie und pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Moritz Jesinghaus
- Institut für Pathologie und pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Katja Steiger
- Institut für Pathologie und pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Wilko Weichert
- Institut für Pathologie und pathologische Anatomie, Technische Universität München, Munich, Germany
- Deutschen Konsortium für Translationale Krebsforschung (DKTK), Partner site Munich, Technische Universität München, Munich, Germany
| | - Helmut Friess
- Chirurgische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Roland Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Hana Algül
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Philipp J. Jost
- Deutschen Konsortium für Translationale Krebsforschung (DKTK), Partner site Munich, Technische Universität München, Munich, Germany
- Innere Medizin III, Hämatologie und Onkologie, Technische Universität München, Munich, Germany
| | - Juliane Ramser
- Klinik und Poliklinik für Frauenheilkunde, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Christine Fischer
- Institut für Humangenetik, Ruprecht-Karls Universität, Heidelberg, Germany
| | - Anne S. Quante
- Klinik und Poliklinik für Frauenheilkunde, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Maximilian Reichert
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Deutschen Konsortium für Translationale Krebsforschung (DKTK), Partner site Munich, Technische Universität München, Munich, Germany
| | - Michael Quante
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Deutschen Konsortium für Translationale Krebsforschung (DKTK), Partner site Munich, Technische Universität München, Munich, Germany
- Klinik für Innere Medizin II, Universität Freiburg, Germany
| |
Collapse
|
11
|
Stevens MA, Rabe KG, Boursi B, Kolluri A, Singh DP, Bamlet WR, Petersen GM. Accuracy of Smoking Status Reporting: Proxy Information in a Rapidly Fatal Cancer Setting. Mayo Clin Proc Innov Qual Outcomes 2020; 4:801-809. [PMID: 33367216 PMCID: PMC7749254 DOI: 10.1016/j.mayocpiqo.2020.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Objective To assess whether patients and relatives can serve as reliable proxy reporters of other family members’ cigarette-smoking history. Patients and Methods Two samples (325 patients, 707 relatives) were identified from the Mayo Clinic Biospecimen Resource for Pancreas Research, enrolled from November, 6, 2000, to March 15, 2018. Smoking-history data, including categorical (ever/never) and quantitative (packs per day and years smoked) smoking measures, were obtained from self-completed questionnaires by patients and relatives. Relative reports were compared with patient reports on self; patient reports were compared with relative reports on self. Results Overall, spouses and first-degree relatives (FDRs) were accurate (94.5%) when reporting patient ever smoking; spouse reports were 98.6% sensitive and 97.7% accurate. Accuracy of patient reports was 97.8% for spouse smoking and 85.5% for FDR smoking; accuracy varied by relationship of FDR. When not concordant, patients generally over-reported daily packs smoked by relatives and under-reported years smoked. Within a 25% agreement range, spouse reports about patients’ daily packs smoked was 46.7%, and years smoked was 69.6%, whereas FDRs were 50% and 64.6%, respectively. When not concordant, relatives generally over-reported daily packs smoked by patients, but no consistent pattern was observed of over- or under-reporting years smoked by patients. Conclusions Patients and relatives can be reliable proxies for smoking history (ever/never) in their family members, especially spouses. An accurate reporting of smoking status will help physicians to better gauge performance status and family smoking exposures to inform disease management.
Collapse
Affiliation(s)
- Maria A Stevens
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN.,Department of Health Policy and Management, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Kari G Rabe
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Ben Boursi
- Department of Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Aarti Kolluri
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Dhruv P Singh
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN.,Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN.,Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
12
|
Jaworski JJ, Morgan RD, Sivakumar S. Circulating Cell-Free Tumour DNA for Early Detection of Pancreatic Cancer. Cancers (Basel) 2020; 12:E3704. [PMID: 33317202 PMCID: PMC7763954 DOI: 10.3390/cancers12123704] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 01/11/2023] Open
Abstract
Pancreatic cancer is a lethal disease, with mortality rates negatively associated with the stage at which the disease is detected. Early detection is therefore critical to improving survival outcomes. A recent focus of research for early detection is the use of circulating cell-free tumour DNA (ctDNA). The detection of ctDNA offers potential as a relatively non-invasive method of diagnosing pancreatic cancer by using genetic sequencing technology to detect tumour-specific mutational signatures in blood samples before symptoms manifest. These technologies are limited by a number of factors that lower sensitivity and specificity, including low levels of detectable ctDNA in early stage disease and contamination with non-cancer circulating cell-free DNA. However, genetic and epigenetic analysis of ctDNA in combination with other standard diagnostic tests may improve early detection rates. In this review, we evaluate the genetic and epigenetic methods under investigation in diagnosing pancreatic cancer and provide a perspective for future developments.
Collapse
Affiliation(s)
- Jedrzej J. Jaworski
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK;
| | - Robert D. Morgan
- Department of Medical Oncology, Christie NHS Foundation Trust, Manchester M20 4BX, UK;
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Shivan Sivakumar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
- Department of Medical Oncology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LE, UK
| |
Collapse
|
13
|
Udgata S, Takenaka N, Bamlet WR, Oberg AL, Yee SS, Carpenter EL, Herman D, Kim J, Petersen GM, Zaret KS. THBS2/CA19-9 Detecting Pancreatic Ductal Adenocarcinoma at Diagnosis Underperforms in Prediagnostic Detection: Implications for Biomarker Advancement. Cancer Prev Res (Phila) 2020; 14:223-232. [PMID: 33067248 DOI: 10.1158/1940-6207.capr-20-0403] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed too late for effective therapy. The classic strategy for early detection biomarker advancement consists of initial retrospective phases of discovery and validation with tissue samples taken from individuals diagnosed with disease, compared with controls. Using this approach, we previously reported the discovery of a blood biomarker panel consisting of thrombospondin-2 (THBS2) and CA19-9 that together could discriminate resectable stage I and IIa PDAC as well as stages III and IV PDAC, with c-statistic values in the range of 0.96 to 0.97 in two phase II studies. We now report that in two studies of blood samples prospectively collected from 1 to 15 years prior to a PDAC diagnosis (Mayo Clinic and PLCO cohorts), THBS2 and/or CA19-9 failed to discriminate cases from healthy controls at the AUC = 0.8 needed. We conclude that PDAC progression may be heterogeneous and for some individuals can be more rapid than generally appreciated. It is important that PDAC early-detection studies incorporate high-risk, prospective prediagnostic cohorts into discovery and validation studies.Prevention Relevance: A blood biomarker panel of THBS2 and CA19-9 detects early stages of pancreatic ductal adenocarcinoma at diagnosis, but not when tested across a population up to 1 year earlier. Our findings suggest serial sampling over time, using prospectively collected samples for biomarker discovery, and more frequent screening of high-risk individuals.
Collapse
Affiliation(s)
- Shirsa Udgata
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, Abramson Cancer Center (Tumor Biology Program), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Naomi Takenaka
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, Abramson Cancer Center (Tumor Biology Program), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - William R Bamlet
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Ann L Oberg
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Stephanie S Yee
- Division of Hematology-Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erica L Carpenter
- Division of Hematology-Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel Herman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jungsun Kim
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, Abramson Cancer Center (Tumor Biology Program), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gloria M Petersen
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, Minnesota.
| | - Kenneth S Zaret
- Institute for Regenerative Medicine, Department of Cell and Developmental Biology, Abramson Cancer Center (Tumor Biology Program), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
14
|
Principe DR, Rana A. Updated risk factors to inform early pancreatic cancer screening and identify high risk patients. Cancer Lett 2020; 485:56-65. [PMID: 32389710 DOI: 10.1016/j.canlet.2020.04.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/06/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic adenocarcinoma (PDAC) is associated with poor clinical outcomes and incomplete responses to conventional therapy. Therefore, there is an unmet clinical need to better understand the predisposing factors for pancreatic cancer in hopes of providing early screening to high-risk patients. While select risk factors such as age, race, and family history, or predisposing syndromes are unavoidable, there are several new and established risk factors that allow for intervention, namely by counseling patients to make the appropriate lifestyle modifications. Here, we discuss the best-studied risk factors for PDAC such as tobacco use and chronic pancreatitis, as well as newly emerging risk factors including select nutritional deficits, bacterial infections, and psychosocial factors. As several of these risk factors appear to be additive or synergistic, by understanding their relationships and offering coordinated, multidisciplinary care to high-risk patients, it may be possible to reduce pancreatic cancer incidence and improve clinical outcomes through early detection.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA; Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Ajay Rana
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
15
|
Primary and Secondary Prevention of Pancreatic Cancer. CURR EPIDEMIOL REP 2019. [DOI: 10.1007/s40471-019-00189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Hamada T, Yuan C, Yurgelun MB, Perez K, Khalaf N, Morales-Oyarvide V, Babic A, Nowak JA, Rubinson DA, Giannakis M, Ng K, Kraft P, Stampfer MJ, Giovannucci EL, Fuchs CS, Ogino S, Wolpin BM. Family history of cancer, Ashkenazi Jewish ancestry, and pancreatic cancer risk. Br J Cancer 2019; 120:848-854. [PMID: 30867564 PMCID: PMC6474278 DOI: 10.1038/s41416-019-0426-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Individuals with a family history of cancer may be at increased risk of pancreatic cancer. Ashkenazi Jewish (AJ) individuals carry increased risk for pancreatic cancer and other cancer types. Methods We examined the association between family history of cancer, AJ heritage, and incident pancreatic cancer in 49 410 male participants of the prospective Health Professionals Follow-up Study. Hazard ratios (HRs) were estimated using multivariable-adjusted Cox proportional hazards models. Results During 1.1 million person-years (1986–2016), 452 participants developed pancreatic cancer. Increased risk of pancreatic cancer was observed in individuals with a family history of pancreatic (HR, 2.79; 95% confidence interval [CI], 1.28–6.07) or breast cancer (HR, 1.40; 95% CI, 1.01–1.94). There was a trend towards higher risk of pancreatic cancer in relation to a family history of colorectal cancer (HR, 1.21; 95% CI, 0.95–1.55) or AJ heritage (HR, 1.29; 95% CI, 0.94–1.77). The risk was highly elevated among AJ men with a family history of breast or colorectal cancer (HR, 2.61 [95% CI, 1.41–4.82] and 1.92 [95% CI, 1.05–3.49], respectively). Conclusion Family history of pancreatic cancer was associated with increased risk of this malignancy. Family history of breast or colorectal cancer was associated with the increased risk among AJ men.
Collapse
Affiliation(s)
- Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Kimberly Perez
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Natalia Khalaf
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Vicente Morales-Oyarvide
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Ana Babic
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Douglas A Rubinson
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA, 02142, USA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Charles S Fuchs
- Yale Cancer Center, 333 Cedar Street, New Haven, CT, 06510, USA.,Department of Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06510, USA.,Smilow Cancer Hospital, 20 York Street, New Haven, CT, 06519, USA
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA.,Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
17
|
Antwi SO, Fagan SE, Chaffee KG, Bamlet WR, Hu C, Polley EC, Hart SN, Shimelis H, Lilyquist J, Gnanaolivu RD, McWilliams RR, Oberg AL, Couch FJ, Petersen GM. Risk of Different Cancers Among First-degree Relatives of Pancreatic Cancer Patients: Influence of Probands' Susceptibility Gene Mutation Status. J Natl Cancer Inst 2019; 111:264-271. [PMID: 29982661 PMCID: PMC6410948 DOI: 10.1093/jnci/djx272] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/01/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Increased risk of malignancies other than pancreatic cancer (PC) has been reported among first-degree relatives (FDRs) of PC patients; however, the roles of susceptibility gene mutations are unclear. We assessed risk for 15 cancers among FDRs of unselected PC probands. METHODS Data on 17 162 FDRs, with more than 336 000 person-years at risk, identified through 2305 sequential PC probands enrolled at Mayo Clinic (2000-2016) were analyzed. Family history data were provided by the probands. Standardized incidence ratios (SIRs) and 95% confidence intervals (CIs) were calculated, comparing malignancies observed among the FDRs with that expected using Surveillance, Epidemiology, and End Results (SEER) data. Genetic testing was performed among a subset of probands (n = 2094), enabling stratified analyses among FDRs based on whether the related proband tested positive or negative for inherited mutation in 22 sequenced cancer susceptibility genes. All statistical tests were two-sided. RESULTS Compared with SEER, PC risk was twofold higher among FDRs of PC probands (SIR = 2.04, 95% CI = 1.78 to 2.31, P < .001). Primary liver cancer risk was elevated among female FDRs (SIR = 2.10, 95% CI = 1.34 to 3.12, P < .001). PC risk was more elevated among FDRs of mutation-positive probands (SIR = 4.32, 95% CI = 3.10 to 5.86) than FDRs of mutation-negative probands (SIR = 1.77, 95% CI = 1.51 to 2.05, between-group P < .001). FDR PC risk was higher when the related proband was younger than age 60 years at diagnosis and mutation-positive (SIR = 5.24, 95% CI = 2.93 to 8.64) than when the proband was younger than age 60 years but mutation-negative (SIR = 1.76, 95% CI = 1.21 to 2.47, between-group P < .001). Breast (SIR = 1.29, 95% CI = 1.01 to 1.63) and ovarian (SIR = 2.38, 95% CI = 1.30 to 4.00) cancers were elevated among FDRs of mutation-positive probands. CONCLUSIONS Our study substantiates twofold risk of PC among FDRs of PC patients and suggests increased risk for primary liver cancer among female FDRs. FDRs of susceptibility mutation carriers had substantially increased risk for PC and increased risk for breast and ovarian cancers.
Collapse
Affiliation(s)
- Samuel O Antwi
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Sarah E Fagan
- Department of Epidemiology, Tulane University, New Orleans, LA
| | - Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - William R Bamlet
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Chunling Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Eric C Polley
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Steven N Hart
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Hermela Shimelis
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Jenna Lilyquist
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | | | - Ann L Oberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Fergus J Couch
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
18
|
Li X, Xu H, Gao P. ABO Blood Group and Diabetes Mellitus Influence the Risk for Pancreatic Cancer in a Population from China. Med Sci Monit 2018; 24:9392-9398. [PMID: 30582832 PMCID: PMC6320638 DOI: 10.12659/msm.913769] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background The mechanism by which diabetes mellitus (DM) impacts the association between ABO blood types and pancreatic cancer is unclear. Material/Methods A retrospective case-control study of 264 patients with pancreatic cancer and 423 age- and sex-matched individuals with nonmalignant diseases was performed to assess whether ABO blood group and DM jointly contribute to pancreatic cancer risk. Results A multivariate analysis with adjustments for risk factors revealed that blood type, chronic pancreatitis, and DM were significantly associated with increased pancreatic cancer risk. The estimated adjusted odds ratios (AORs with 95% confidence intervals [CIs]) were 2.130 (1.409–3.220) for blood type A, 2.383 (1.313–4.325) for blood type AB, 1.518 (1.012–2.276) for DM, and 10.930 (1.202–99.405) for chronic pancreatitis. Blood type A significantly modified the risk for pancreatic cancer in individuals with DM (AOR, 3.506; 95% CI, 1.659–7.409). Conclusions The risk for pancreatic cancer was associated with ABO blood type, DM, and chronic pancreatitis in a Chinese population. The risk was greatest for individuals with blood type A and DM.
Collapse
Affiliation(s)
- Xu Li
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China (mainland)
| | - Hongqin Xu
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China (mainland).,Jilin Province Key Laboratory of Infectious Disease, Laboratory of Molecular Virology, Changchun, Jilin, China (mainland)
| | - Pujun Gao
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
19
|
McWilliams RR, Wieben ED, Chaffee KG, Antwi SO, Raskin L, Olopade OI, Li D, Highsmith WE, Colon-Otero G, Khanna LG, Permuth JB, Olson JE, Frucht H, Genkinger J, Zheng W, Blot WJ, Wu L, Almada LL, Fernandez-Zapico ME, Sicotte H, Pedersen KS, Petersen GM. CDKN2A Germline Rare Coding Variants and Risk of Pancreatic Cancer in Minority Populations. Cancer Epidemiol Biomarkers Prev 2018; 27:1364-1370. [PMID: 30038052 PMCID: PMC6214745 DOI: 10.1158/1055-9965.epi-17-1065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/13/2018] [Accepted: 07/11/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Pathogenic germline mutations in the CDKN2A tumor suppressor gene are rare and associated with highly penetrant familial melanoma and pancreatic cancer in non-Hispanic whites (NHW). To date, the prevalence and impact of CDKN2A rare coding variants (RCV) in racial minority groups remain poorly characterized. We examined the role of CDKN2A RCVs on the risk of pancreatic cancer among minority subjects.Methods: We sequenced CDKN2A in 220 African American (AA) pancreatic cancer cases, 900 noncancer AA controls, and 183 Nigerian controls. RCV frequencies were determined for each group and compared with that of 1,537 NHW patients with pancreatic cancer. Odds ratios (OR) and 95% confidence intervals (CI) were calculated for both a case-case comparison of RCV frequencies in AAs versus NHWs, and case-control comparison between AA cases versus noncancer AA controls plus Nigerian controls. Smaller sets of Hispanic and Native American cases and controls also were sequenced.Results: One novel missense RCV and one novel frameshift RCV were found among AA patients: 400G>A and 258_278del. RCV carrier status was associated with increased risk of pancreatic cancer among AA cases (11/220; OR, 3.3; 95% CI, 1.5-7.1; P = 0.004) compared with AA and Nigerian controls (17/1,083). Further, AA cases had higher frequency of RCVs: 5.0% (OR, 13.4; 95% CI, 4.9-36.7; P < 0.001) compared with NHW cases (0.4%).Conclusions: CDKN2A RCVs are more common in AA than in NHW patients with pancreatic cancer and associated with moderately increased pancreatic cancer risk among AAs.Impact: RCVs in CDKN2A are frequent in AAs and are associated with risk for pancreatic cancer. Cancer Epidemiol Biomarkers Prev; 27(11); 1364-70. ©2018 AACR.
Collapse
Affiliation(s)
| | - Eric D Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kari G Chaffee
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Samuel O Antwi
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida
| | - Leon Raskin
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Olufunmilayo I Olopade
- Departments of Medicine and Human Genetics, University of Chicago Medical Center, Chicago, Illinois
| | - Donghui Li
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - W Edward Highsmith
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Gerardo Colon-Otero
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida
| | - Lauren G Khanna
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jennifer B Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Harold Frucht
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Jeanine Genkinger
- Department of Epidemiology, Columbia University Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Wei Zheng
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - William J Blot
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Lang Wu
- Division of Epidemiology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Luciana L Almada
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota
| | - Martin E Fernandez-Zapico
- Schulze Center for Novel Therapeutics, Division of Oncology Research, Mayo Clinic, Rochester, Minnesota
| | - Hugues Sicotte
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
20
|
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: 5.0] [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
|
21
|
Jones JK, Tave A, Pezzullo JC, Kardia S, Lippes J. Long-term risk of hysterectomy and ectopic pregnancy among Vietnamese women using the quinacrine hydrochloride pellet system vs. intrauterine devices or tubal ligation for contraception. EUR J CONTRACEP REPR 2018; 23:105-115. [PMID: 29683010 DOI: 10.1080/13625187.2018.1449823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES Determine the long-term risk of hysterectomy and ectopic pregnancy in women using the quinacrine hydrochloride pellet system of permanent contraception (QS) relative to the comparable risk in women using Copper T intrauterine device (IUD) or tubal ligation surgery (TL) for long-term or permanent contraception. METHODS This was a retrospective cohort study, conducted in the Northern Vietnamese provinces of Ha Nam, Nam Dinh, Ninh Binh and Thai Binh. Women who had their first QS procedure, last IUD insertion or TL between 1989 and 1996 were interviewed regarding post-procedure health outcomes approximately 16 years post exposure. RESULTS A 95% response rate resulted in 21,040 completed interviews. Overall incidence rates were low for both outcomes (91/100,000 women years of follow-up and 22/100,000 women years of follow-up for hysterectomy and ectopic pregnancy, respectively). After accounting for variations in baseline characteristics between women choosing QS vs. the other two contraceptive methods, no significant excess hazard of either hysterectomy or ectopic pregnancy was associated with QS. CONCLUSIONS No significant excess long-term risk of hysterectomy or ectopic pregnancy was found among a large group of women using QS vs. IUD or TL for contraception after an average 16 years of follow-up.
Collapse
Affiliation(s)
| | | | - John C Pezzullo
- b Department of Biomathematics and Biostatistics , Georgetown University , Washington , DC , USA
| | - Sharon Kardia
- c School of Public Health , University of Michigan , Ann Arbor , MI , USA
| | - Jack Lippes
- d School of Medicine , State University of New York at Buffalo , Buffalo , NY , USA
| |
Collapse
|
22
|
Sohal DPS, Willingham FF, Falconi M, Raphael KL, Crippa S. Pancreatic Adenocarcinoma: Improving Prevention and Survivorship. Am Soc Clin Oncol Educ Book 2017; 37:301-310. [PMID: 28561672 DOI: 10.1200/edbk_175222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Pancreatic cancer is a growing problem in oncology, given slowly rising incidence and continued suboptimal outcomes. A concerted effort to reverse this tide will require prevention, early diagnosis, and improved systemic therapy for curable disease. We focus on these aspects in detail in this study. Hereditary pancreatic cancer is an underappreciated area. With the growing use of genomics (both somatic and germline) in cancer care, there is increasing recognition of hereditary pancreatic cancer cases: around 10% of all pancreatic cancer may be related to familial syndromes, such as familial atypical multiple mole and melanoma (FAMMM) syndrome, hereditary breast and ovarian cancer, Lynch syndrome, and Peutz-Jeghers syndrome. Screening and surveillance guidelines by various expert groups are discussed. Management of resectable pancreatic cancer is evolving; the use of multiagent systemic therapies, in the adjuvant and neoadjuvant settings, is discussed. Current and emerging data, along with ongoing clinical trials addressing important questions in this area, are described. Surveillance recommendations based on latest ASCO guidelines are also discussed. Finally, the multimodality management of borderline resectable pancreatic cancer is discussed. The various clinicoanatomic definitions of this entity, followed by consensus definitions, are described. Then, we focus on current opinions and practices around neoadjuvant therapy, discussing chemotherapy and radiation aspects, and the role of surgical resection.
Collapse
Affiliation(s)
- Davendra P S Sohal
- From the Cleveland Clinic, Cleveland, OH; Emory University School of Medicine, Atlanta, GA; Division of Pancreatic Surgery, Università Vita-Salute, San Raffaele Scientific Institute, Milan, Italy
| | - Field F Willingham
- From the Cleveland Clinic, Cleveland, OH; Emory University School of Medicine, Atlanta, GA; Division of Pancreatic Surgery, Università Vita-Salute, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Falconi
- From the Cleveland Clinic, Cleveland, OH; Emory University School of Medicine, Atlanta, GA; Division of Pancreatic Surgery, Università Vita-Salute, San Raffaele Scientific Institute, Milan, Italy
| | - Kara L Raphael
- From the Cleveland Clinic, Cleveland, OH; Emory University School of Medicine, Atlanta, GA; Division of Pancreatic Surgery, Università Vita-Salute, San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- From the Cleveland Clinic, Cleveland, OH; Emory University School of Medicine, Atlanta, GA; Division of Pancreatic Surgery, Università Vita-Salute, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
23
|
Association between family cancer history and risk of pancreatic cancer. Cancer Epidemiol 2016; 45:145-150. [PMID: 27810486 DOI: 10.1016/j.canep.2016.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/21/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022]
Abstract
PURPOSE Family history of pancreatic adenocarcinoma is an established risk factor for the disease. However, associations of pancreatic cancer with other familial cancers are less clear. We analyzed data from the Queensland Pancreatic Cancer Study (QPCS), an Australian population-based case-control study, to investigate associations between family history of various cancer types and risk of pancreatic cancer. MATERIALS AND METHODS Our study included 591 pancreatic cancer patients and 646 controls, all of whom self-reported the histories of cancer in their first-degree relatives. We used logistic regression to estimate adjusted odds ratios (ORs) and their 95% confidence intervals (CIs). Based on our results, we conducted a systematic literature review using the Medline (OVID) database to identify articles pertaining to the association between family history of melanoma and risk of pancreatic cancer. A meta-analysis including associations in five published studies, unpublished results from a study co-author and the QPCS results was then performed using the DerSimonian and Laird random-effects model. RESULTS Cases were more likely than controls to report a family history of pancreatic cancer (OR 2.20, 95% CI 1.16-4.19) and melanoma (OR 1.74, 95% CI 1.03-2.95), but not of breast, ovarian, respiratory, other gastrointestinal or prostate cancer. Meta-analysis of melanoma family history and pancreatic cancer risk yielded an OR of 1.22 (95% CI 1.00-1.51). CONCLUSIONS Our results yield further evidence of increased risk of pancreatic cancer in those with family histories of the disease. We also provide suggestive evidence of an association between family history of melanoma and risk of pancreatic cancer.
Collapse
|
24
|
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
|
25
|
Antwi SO, Oberg AL, Shivappa N, Bamlet WR, Chaffee KG, Steck SE, Hébert JR, Petersen GM. Pancreatic cancer: associations of inflammatory potential of diet, cigarette smoking and long-standing diabetes. Carcinogenesis 2016; 37:481-90. [PMID: 26905587 PMCID: PMC4843052 DOI: 10.1093/carcin/bgw022] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/04/2016] [Accepted: 02/11/2016] [Indexed: 01/02/2023] Open
Abstract
Epidemiologic studies show strong associations between pancreatic cancer (PC) and inflammatory stimuli or conditions such as cigarette smoking and diabetes, suggesting that inflammation may play a key role in PC. Studies of dietary patterns and cancer outcomes also suggest that diet might influence an individual's risk of PC by modulating inflammation. We therefore examined independent and joint associations between inflammatory potential of diet, cigarette smoking and long-standing (≥5 years) type II diabetes in relation to risk of PC. Analyses included data from 817 cases and 1756 controls. Inflammatory potential of diet was measured using the dietary inflammatory index (DII), calculated from dietary intake assessed via a 144-item food frequency questionnaire, and adjusted for energy intake. Information on smoking and diabetes were obtained via risk factor questionnaires. Associations were examined using multivariable-adjusted logistic regression. Higher DII scores, reflecting a more proinflammatory diet, were associated with increased risk of PC [odds ratio (OR)Quintile 5 versus 1 = 2.54, 95% confidence interval (CI) = 1.87-3.46, P trend < 0.0001]. Excess risk of PC also was observed among former (OR = 1.29, 95% CI = 1.07-1.54) and current (OR = 3.40, 95% CI = 2.28-5.07) smokers compared with never smokers, and among participants with long-standing diabetes (OR = 3.09, 95% CI = 2.02-4.72) compared with nondiabetics. Joint associations were observed for the combined effects of having greater than median DII score, and being a current smoker (OR = 4.79, 95% CI = 3.00-7.65) or having long-standing diabetes (OR = 6.03, 95% CI = 3.41-10.85). These findings suggest that a proinflammatory diet may act as cofactor with cigarette smoking and diabetes to increase risk of PC beyond the risk of any of these factors alone.
Collapse
Affiliation(s)
- Samuel O. Antwi
- Division of Epidemiology and
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Ann L. Oberg
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
| | - Nitin Shivappa
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - William R. Bamlet
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
| | - Kari G. Chaffee
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Charlton 6-243, Rochester, MN 55905, USA and
| | - Susan E. Steck
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - James R. Hébert
- Cancer Prevention and Control Program and
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Gloria M. Petersen
- *To whom correspondence should be addressed. Tel: +1 5075 381563; Fax: +1 5072 662478;
| |
Collapse
|
26
|
Abstract
Familial pancreatic cancer (FPC) kindreds have at least 2 first-degree relatives with pancreatic ductal adenocarcinoma. Studies of FPC have focused on the discovery of genetic cause and on the management of those at genetically high risk. Research reveals that a half dozen known hereditary syndromes or genes are associated with increased risk of developing pancreatic cancer, the most prominent of which are BRCA2 and CDKN2A. Genetic risk assessment and testing is already available. Owing to limited experience worldwide, guidance is often based on expert opinion, although all agree that research is needed to improve the shaping of options.
Collapse
Affiliation(s)
- Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic Cancer Center, Mayo Clinic, Charlton 6-243, Rochester, MN 55905, USA.
| |
Collapse
|
27
|
Smith AL, Bascuñana C, Hall A, Salman A, Andrei AZ, Volenik A, Rothenmund H, Ferland D, Lamoussenery D, Kamath AS, Amre R, Caglar D, Gao ZH, Haegert DG, Kanber Y, Michel RP, Omeroglu-Altinel G, Asselah J, Bouganim N, Kavan P, Arena G, Barkun J, Chaudhury P, Gallinger S, Foulkes WD, Omeroglu A, Metrakos P, Zogopoulos G. Establishing a clinic-based pancreatic cancer and periampullary tumour research registry in Quebec. ACTA ACUST UNITED AC 2015; 22:113-21. [PMID: 25908910 DOI: 10.3747/co.22.2300] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Enrolling patients in studies of pancreatic ductal adenocarcinoma (pdac) is challenging because of the high fatality of the disease. We hypothesized that a prospective clinic-based study with rapid ascertainment would result in high participation rates. Using that strategy, we established the Quebec Pancreas Cancer Study (qpcs) to investigate the genetics and causes of pdac and other periampullary tumours (pats) that are also rare and underrepresented in research studies. METHODS Patients diagnosed with pdac or pat were introduced to the study at their initial clinical encounter, with a strategy to enrol participants within 2 weeks of diagnosis. Patient self-referrals and referrals of unaffected individuals with an increased risk of pdac were also accepted. Family histories, epidemiologic and clinical data, and biospecimens were collected. Additional relatives were enrolled in families at increased genetic risk. RESULTS The first 346 completed referrals led to 306 probands being enrolled, including 190 probands affected with pdac, who represent the population focus of the qpcs. Participation rates were 88.4% for all referrals and 89.2% for pdac referrals. Family history, epidemiologic and clinical data, and biospecimens were ascertained from 91.9%, 54.6%, and 97.5% respectively of patients with pdac. Although demographics and trends in risk factors in our patients were consistent with published statistics for patients with pdac, the qpcs is enriched for families with French-Canadian ancestry (37.4%), a population with recurrent germ-line mutations in hereditary diseases. CONCLUSIONS Using rapid ascertainment, a pdac and pat research registry with high participation rates can be established. The qpcs is a valuable research resource and its enrichment with patients of French-Canadian ancestry provides a unique opportunity for studies of heredity in these diseases.
Collapse
Affiliation(s)
- A L Smith
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - C Bascuñana
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - A Hall
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - A Salman
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - A Z Andrei
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC
| | - A Volenik
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| | - H Rothenmund
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| | - D Ferland
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - D Lamoussenery
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - A S Kamath
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - R Amre
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - D Caglar
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - Z H Gao
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - D G Haegert
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - Y Kanber
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - R P Michel
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | | | - J Asselah
- Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - N Bouganim
- Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - P Kavan
- Hepato-Pancreato-Biliary Oncology, McGill University Health Centre, Montreal, QC
| | - G Arena
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - J Barkun
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - P Chaudhury
- Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - S Gallinger
- The Research Institute of the McGill University Health Centre, Montreal, QC
| | - W D Foulkes
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| | - A Omeroglu
- Department of Pathology, McGill University Health Centre, Montreal, QC
| | - P Metrakos
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC
| | - G Zogopoulos
- The Research Institute of the McGill University Health Centre, Montreal, QC. ; The Goodman Cancer Research Centre, McGill University, Montreal, QC. ; Hepato-Pancreato-Biliary and Transplant Surgery, McGill University Health Centre, Montreal, QC. ; Program in Cancer Genetics, Department of Oncology and Human Genetics, McGill University, Montreal, QC
| |
Collapse
|
28
|
Li J, Zhang JT, Jiang X, Shi X, Shen J, Feng F, Chen J, Liu G, He P, Jiang J, Tsang LL, Wang Y, Rosell R, Jiang L, He J, Chan HC. The cystic fibrosis transmembrane conductance regulator as a biomarker in non-small cell lung cancer. Int J Oncol 2015; 46:2107-15. [PMID: 25760446 DOI: 10.3892/ijo.2015.2921] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 01/12/2015] [Indexed: 01/12/2023] Open
Abstract
An increased risk of non-small cell lung cancer (NSCLC) in cystic fibrosis (CF) patients and carriers of CF transmembrane conductance regulator (CFTR) mutations has been proposed. However, the role of CFTR in lung cancer remains controversial. In the present study, CFTR expression was assessed in 165 NSCLC tumors and 22 normal lung samples with validation in an independent series of 131 samples. The effect of gain and loss of CFTR on the malignant behavior of NSCLC was examined. The effect of CFTR manipulation on tumor metastasis was examined in a mouse model. Expression of CFTR was downregulated in NSCLC (p=0.041). Low CFTR expression was correlated with advanced stage (p<0.001) and lymph node metastasis (p=0.009). Low CFTR expression was significantly associated with poor prognosis (overall survival: 45 vs. 36 months, p<0.0001; progression-free survival: 41 vs. 30 months, p=0.007). Knockdown of CFTR in NSCLC cells enhanced malignant behavior (epithelial-mesenchymal transition, invasion and migration); in contrast, overexpression of CFTR suppressed cancer progression in vitro and in vivo. The tumor-suppressing effect of CFTR was associated with inhibition of multiple uPA/uPAR-mediated malignant traits in culture. These results show that CFTR plays a role in inhibition of NSCLC metastasis and suggest that CFTR may serve as a novel indicator for predicting adverse prognosis and metastasis in NSCLC patients.
Collapse
Affiliation(s)
- Jin Li
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Jie Ting Zhang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, P.R. China
| | - Xiaohua Jiang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, P.R. China
| | - Xiaoshun Shi
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Jianfei Shen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Fenglan Feng
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Jingyi Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Guihong Liu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Ping He
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Juhong Jiang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Lai Ling Tsang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, P.R. China
| | - Yan Wang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, P.R. China
| | - Rafael Rosell
- Catalan Institute of Oncology, Badalona, Catalonia, Spain
| | - Long Jiang
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Jianxing He
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, P.R. China
| |
Collapse
|
29
|
Tehranifar P, Wu HC, Shriver T, Cloud AJ, Terry MB. Validation of family cancer history data in high-risk families: the influence of cancer site, ethnicity, kinship degree, and multiple family reporters. Am J Epidemiol 2015; 181:204-12. [PMID: 25568166 DOI: 10.1093/aje/kwu258] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Information on family cancer history (FCH) is often collected for first-degree relatives, but more extensive FCH information is critical for greater accuracy in risk assessment. Using self-reported diagnosis of cancer as the gold standard, we examined differences in the sensitivity and specificity of relative-reported FCH by cancer site, race/ethnicity, language preference, and kinship degree (1,524 individuals from 557 families; average number of relatives per family = 2.7). We evaluated the impact of FCH data collected in 2007-2013 from multiple relatives by comparing mean values and proportions for the number of relatives with any cancer, breast cancer, or ovarian cancer as reported by a single relative and by multiple relatives in the same family. The sensitivity of FCH was lower in Hispanics, Spanish-speaking persons, and third-degree relatives (e.g., for all cancers, sensitivities were 80.7%, 87.4%, and 91.0% for third-, second-, and first-degree relatives, respectively). FCH reported by multiple relatives included a higher number of relatives with cancer than the number reported by a single relative (e.g., mean increase of 1.2 relatives with any cancer), with more relatives diagnosed with any cancer, breast cancer, and ovarian cancer in 52%, 36% and 12% of families, respectively. Collection of FCH data from multiple relatives may provide a more comprehensive picture of FCH and may potentially improve risk assessment and preventive care.
Collapse
|
30
|
Milne R, La Vecchia C, Van Steen K, Hahn S, Buchholz M, Costello E, Esposito I, Hoheisel JD, Lange B, Lopez-Bigas N, Michalski CW, Real FX, Brand A, Malats N. EU Pancreas: an integrated European platform for pancreas cancer research--from basic science to clinical and public health interventions for a rare disease. Public Health Genomics 2014; 16:305-12. [PMID: 24503591 DOI: 10.1159/000355937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Large-scale international collaboration is essential to decipher relevant information in the context of omics-scale interrogations in cancer research. This is even more important for rare and fatal diseases like pancreas cancer (PC). METHODS The COST Action BM1204 is a unique platform to facilitate the collaboration of a broad range of European and international PC multidisciplinary research groups in order to: (1) integrate knowledge and experience in a multidisciplinary way 'from cell to society', (2) promote the application of uniform study tools and protocols, (3) foster their optimal use by early-stage researchers, (4) enhance the mobility and training of researchers, and (5) disseminate the results produced to the broader society. RESULTS This Action will develop novel interdisciplinary tools for collaborative research to improve our understanding of PC and its prevention, diagnosis and treatment. It also aims to answer questions related to the etiology, early detection, evidence-based and personalized treatment, and health management for PC. Furthermore, the Action will contribute to new insights into PC personalized medicine and beyond as well as to the understanding of complex and rare diseases taking PC as a best practice example. The Action aims at attracting young scholars across a range of disciplines in collaboration with more experienced researchers and enhancing active European participation in the international scenario of PC research. CONCLUSION The ultimate aim is to foster PC research in Europe and to coordinate this effort with other international initiatives to reduce disease mortality.
Collapse
Affiliation(s)
- R Milne
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhang JT, Jiang XH, Xie C, Cheng H, Da Dong J, Wang Y, Fok KL, Zhang XH, Sun TT, Tsang LL, Chen H, Sun XJ, Chung YW, Cai ZM, Jiang WG, Chan HC. Downregulation of CFTR promotes epithelial-to-mesenchymal transition and is associated with poor prognosis of breast cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2961-2969. [PMID: 23916755 DOI: 10.1016/j.bbamcr.2013.07.021] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 07/22/2013] [Accepted: 07/24/2013] [Indexed: 01/12/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT), a process involving the breakdown of cell-cell junctions and loss of epithelial polarity, is closely related to cancer development and metastatic progression. While the cystic fibrosis transmembrane conductance regulator (CFTR), a Cl(-) and HCO3(-) conducting anion channel expressed in a wide variety of epithelial cells, has been implicated in the regulation of epithelial polarity, the exact role of CFTR in the pathogenesis of cancer and its possible involvement in EMT process have not been elucidated. Here we report that interfering with CFTR function either by its specific inhibitor or lentiviral miRNA-mediated knockdown mimics TGF-β1-induced EMT and enhances cell migration and invasion in MCF-7. Ectopic overexpression of CFTR in a highly metastatic MDA-231 breast cancer cell line downregulates EMT markers and suppresses cell invasion and migration in vitro, as well as metastasis in vivo. The EMT-suppressing effect of CFTR is found to be associated with its ability to inhibit NFκB targeting urokinase-type plasminogen activator (uPA), known to be involved in the regulation of EMT. More importantly, CFTR expression is found significantly downregulated in primary human breast cancer samples, and is closely associated with poor prognosis in different cohorts of breast cancer patients. Taken together, the present study has demonstrated a previously undefined role of CFTR as an EMT suppressor and its potential as a prognostic indicator in breast cancer.
Collapse
Affiliation(s)
- Jie Ting Zhang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiao Hua Jiang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong; Key Laboratory for Regenerative Medicine, Ji Nan University-The Chinese University of Hong Kong, Ministry of Education of The People's Republic of China, China
| | - Chen Xie
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hong Cheng
- Department of Pathology, State Key Laboratory of Cancer Biology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jian Da Dong
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yan Wang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kin Lam Fok
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Xiao Hu Zhang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ting Ting Sun
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lai Ling Tsang
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hao Chen
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong; Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xiao Juan Sun
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yiu Wa Chung
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhi Ming Cai
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wen Guo Jiang
- Metastasis and Angiogenesis Research Group, Department of Surgery, Cardiff University School of Medicine, Cardiff, UK.
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong; Key Laboratory for Regenerative Medicine, Ji Nan University-The Chinese University of Hong Kong, Ministry of Education of The People's Republic of China, China; Sichuan University-The Chinese University of Hong Kong Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Chengdu, China.
| |
Collapse
|
32
|
Survival is associated with genetic variation in inflammatory pathway genes among patients with resected and unresected pancreatic cancer. Ann Surg 2013; 257:1096-102. [PMID: 23360921 DOI: 10.1097/sla.0b013e318275b7e5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To test whether or not the association between inflammation and pancreatic ductal adenocarcinoma (PC) is facilitated by host susceptibility, specifically by genetic polymorphisms in inflammation-related genes. SUMMARY BACKGROUND DATA Inflammation has been linked to PC. Reports have cited an increased expression of proinflammatory mediators, such as NF-κB and COX, in PC but not in normal adjacent tissue, suggesting a possible role in carcinogenesis. We sought to further understand the role that genetic variants in the NF-κB inflammatory pathway play in the development and progression of PC. METHODS We genotyped 1536 tag single nucleotide polymorphisms (SNPs) in 102 candidate genes of multiple inflammatory pathways in 1308 white patients with PC who were divided into 3 groups on the basis of the extent of disease: resected for cure (n = 400), locally advanced/unresected (n = 443), and metastatic (n = 465). Survival analysis was performed using Kaplan-Meier curves and Cox proportional hazards regression models. Statistical significance was set at less than 0.001 to control for multiple testing. RESULTS Median age was 67 (28.0-91.0) years, and 57% were men. Median survival for each of the 3 groups (resected, locally advanced, and metastatic) was 23.7, 9.4, and 6.6 months, respectively (P < 0.0001). In the resected group, carriers of a minor allele for either rs3824872 (MAPK8IP1) or rs8064821 (SOCS3) were associated with a 10- and 6-month survival advantage compared with noncarriers in patients with resected disease, with an additional 2-year survival if both minor alleles were present. With locally advanced disease, SNP rs1124736 (IGF1R) was associated with improved survival if they had a copy of the G allele, hazard ratio of 0.57 (95% confidence interval: 0.42-0.77); P = 0.0002. In addition, 4 SNPs in patients with metastatic disease were found to be associated with worse survival and 2 associated with improved overall survival, but the differences in survival were deemed not clinically significant. CONCLUSIONS SNPs in the inflammatory pathway genes MAPK8IP1 and SOCS3 were associated with increased overall survival in patients undergoing potentially curative resection and may be used in the future as markers to predict survival. Future research is needed to determine the functional relevance of these loci.
Collapse
|
33
|
Wang DS, Wang ZQ, Zhang L, Qiu MZ, Luo HY, Ren C, Zhang DS, Wang FH, Li YH, Xu RH. Are risk factors associated with outcomes in pancreatic cancer? PLoS One 2012; 7:e41984. [PMID: 22911869 PMCID: PMC3404018 DOI: 10.1371/journal.pone.0041984] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 06/27/2012] [Indexed: 02/08/2023] Open
Abstract
Background The development of pancreatic cancer is a process in which genes interact with environmental factors. We performed this study to determine the effects of the ABO blood group, obesity, diabetes mellitus, metabolic syndrome (MetS), smoking, alcohol consumption and hepatitis B viral (HBV) infection on patient survival. Methods A total of 488 patients with pancreatic cancer were evaluated. Result Patients who presented as chronic carriers of HBV infection were younger at disease onset (p = 0.001) and more predominantly male (p = 0.020) than those never exposed to HBV. Patients with MetS had later disease staging (p = 0.000) and a lower degree of pathological differentiation (p = 0.008) than those without MetS. In a univariate analysis, the ABO blood group, smoking and alcohol consumption were not associated with overall survival. HBsAg–positivity and elevated fasting plasma glucose were significantly associated with unfavorable survival though not in the multivariate analysis. The presence of MetS (HR: 1.541, 95% CI: 1.095–2.169, p = 0.013), age ≥65, an elevated CA19–9 baseline level, TNM staging, the type of surgery, the degree of differentiation and chemotherapy were independently associated with overall survival. Conclusion We report, for the first time, that patients with chronic HBV infection may represent a special subtype of pancreatic cancer, who have a younger age of disease onset and male dominancy. Patients with MetS had later disease staging and a poorer histological grade. Patients with MetS demonstrated significantly poorer survival.
Collapse
Affiliation(s)
- De-shen Wang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-qiang Wang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Le Zhang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Miao-zhen Qiu
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui-yan Luo
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chao Ren
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dong-sheng Zhang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feng-hua Wang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yu-hong Li
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Rui-hua Xu
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- * E-mail:
| |
Collapse
|
34
|
Xie C, Jiang XH, Zhang JT, Sun TT, Dong JD, Sanders AJ, Diao RY, Wang Y, Fok KL, Tsang LL, Yu MK, Zhang XH, Chung YW, Ye L, Zhao MY, Guo JH, Xiao ZJ, Lan HY, Ng CF, Lau KM, Cai ZM, Jiang WG, Chan HC. CFTR suppresses tumor progression through miR-193b targeting urokinase plasminogen activator (uPA) in prostate cancer. Oncogene 2012; 32:2282-91, 2291.e1-7. [PMID: 22797075 DOI: 10.1038/onc.2012.251] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cystic fibrosis (CF) transmembrane conductance regulator (CFTR) is expressed in the epithelial cells of a wide range of organs/tissues from which most cancers are derived. Although accumulating reports have indicated the association of cancer incidence with genetic variations in CFTR gene, the exact role of CFTR in cancer development and the possible underlying mechanism have not been elucidated. Here, we report that CFTR expression is significantly decreased in both prostate cancer cell lines and human prostate cancer tissue samples. Overexpression of CFTR in prostate cancer cell lines suppresses tumor progression (cell growth, adhesion and migration), whereas knockdown of CFTR leads to enhanced malignancies both in vitro and in vivo. In addition, we demonstrate that CFTR knockdown-enhanced cell proliferation, cell invasion and migration are significantly reversed by antibodies against either urokinase plasminogen activator (uPA) or uPA receptor (uPAR), which are known to be involved in various malignant traits of cancer development. More interestingly, overexpression of CFTR suppresses uPA by upregulating the recently described tumor suppressor microRNA-193b (miR-193b), and overexpression of pre-miR-193b significantly reverses CFTR knockdown-enhanced malignant phenotype and abrogates elevated uPA activity in prostate cancer cell line. Finally, we show that CFTR gene transfer results in significant tumor repression in prostate cancer xenografts in vivo. Taken together, the present study has demonstrated a previously undefined tumor-suppressing role of CFTR and its involvement in regulation of miR-193b in prostate cancer development.
Collapse
Affiliation(s)
- C Xie
- Epithelial Cell Biology Research Center, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
OBJECTIVES To investigate the incidence, characteristics, and prognostic impact of prior extrapancreatic malignancies on patients with pancreatic cancer (PDAC). METHODS Records from 1733 patients who underwent surgery for PDAC were analyzed for the occurrence of prior extrapancreatic malignancies. Patients' records showing extrapancreatic malignancies were then analyzed for tumor type, epidemiological data, risk factors, PDAC tumor stage, and long-term survival. RESULTS A total of 239 patients with PDAC (13.8%) had a history of 271 extrapancreatic tumors; 26 patients had a history of two pancreatic cancers, and 3 patients had 3 extrapancreatic cancers. The most common extrapancreatic tumors were breast cancer (56 patients) and prostate cancer (41 patients), followed by colorectal, reno/urothelial, and gynecologic tumors (39, 32, and 23 patients, respectively). No significant difference in overall survival was found between patients with PDAC with or without extrapancreatic malignancies. CONCLUSIONS Pancreatic cancer is associated with extrapancreatic malignancies in a remarkable number of patients. A history of extrapancreatic malignancies does not influence prognosis and should not be an obstacle to a curative therapeutic approach. Surveillance of patients with extrapancreatic malignancies, especially breast, prostate, and colorectal cancer, could allow for earlier PDAC diagnosis and therefore improve prognosis of these patients.
Collapse
|
36
|
Amin S, McBride R, Kline J, Mitchel EB, Lucas AL, Neugut AI, Frucht H. Incidence of subsequent pancreatic adenocarcinoma in patients with a history of nonpancreatic primary cancers. Cancer 2012; 118:1244-51. [PMID: 21887676 PMCID: PMC3677019 DOI: 10.1002/cncr.26414] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Revised: 05/31/2011] [Accepted: 06/06/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Several environmental risk factors are known to predispose individuals to pancreatic cancer, and up to 15% of pancreatic cancers have an inherited component. Understanding metachronous cancer associations can modify pancreas cancer risk. The objective of this study was to investigate the association of nonpancreatic cancers with subsequent pancreatic adenocarcinoma. METHODS The authors used data from the US Surveillance, Epidemiology, and End Results (SEER) registries to identify 1,618,834 individuals who had a primary malignancy and subsequent pancreatic adenocarcinoma (n = 4013). Standardized incidence ratios were calculated as an approximation of relative risk (RR) for the occurrence of pancreatic adenocarcinoma after another primary malignancy. RESULTS Among patients who were diagnosed with a first primary malignancy at ages 20 to 49 years, the risk of subsequent pancreatic adenocarcinoma was increased among patients who had cancers of the ascending colon (relative risk [RR], 4.62; 95% confidence interval [CI], 1.86-9.52), hepatic flexure (RR, 5.42; 95% CI, 1.12-15.84), biliary system (RR, 13.14; 95% CI, 4.27-30.66), breast (RR, 1.32; 95% CI, 1.09-1.59), uterine cervix (RR, 1.61; 95% CI, 1.02-2.41), testes (RR, 2.78; 95% CI, 1.83-4.05), and hematopoietic system (RR, 1.83; 95% CI, 1.28-2.53). Among patients who had a first malignancy at ages 50 to 64 years, the risk was increased after cancers of the stomach (RR, 1.88; 95% CI, 1.13-2.93), hepatic flexure (RR, 2.25; 95% CI, 1.08-4.13), lung and bronchus (RR, 1.46; 95% CI, 1.16-1.82), pharynx (RR, 2.26; 95% CI, 1.13-4.04), and bladder (RR, 1.24; 95% CI, 1.03-1.48). Among patients who had a primary cancer after age 65 years, the risk was increased after cancers of the stomach (RR, 1.79; 95% CI, 1.23-2.53), hepatic flexure (RR, 1.76; 95% CI, 1.06-2.75), biliary system (RR, 2.35; 95% CI, 1.17-4.20), and uterus (RR, 1.23; 95% CI, 1.03-1.47). CONCLUSIONS The results from the current population-based data set suggested that pancreatic adenocarcinoma is associated with certain primary cancers. Genetic predisposition and common environmental and behavioral risk factors all may contribute to this observation. Specific tumor associations will guide future risk-stratification efforts.
Collapse
Affiliation(s)
- Sunil Amin
- College of Physicians and Surgeons, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Muzzi Mirza Pancreatic Cancer Prevention & Genetics Program, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
| | - Russell McBride
- Department of Epidemiology, Mailman School of Public Health
- Herbert Irving Comprehensive Cancer Center
| | - Jennie Kline
- Sergievsky Center, Columbia University, New York, NY and the New York State Psychiatric Institute, New York, NY
- Department of Epidemiology, Mailman School of Public Health
| | - Elana B. Mitchel
- College of Physicians and Surgeons, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Muzzi Mirza Pancreatic Cancer Prevention & Genetics Program, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
| | - Aimee L. Lucas
- Muzzi Mirza Pancreatic Cancer Prevention & Genetics Program, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Division of Digestive and Liver Diseases, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Department of Medicine, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
| | - Alfred I. Neugut
- Department of Medicine, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Herbert Irving Comprehensive Cancer Center
| | - Harold Frucht
- Muzzi Mirza Pancreatic Cancer Prevention & Genetics Program, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Division of Digestive and Liver Diseases, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Department of Medicine, Columbia University, New York, NY and New York Presbyterian Hospital, New York, NY
- Herbert Irving Comprehensive Cancer Center
| |
Collapse
|
37
|
Wang DS, Chen DL, Ren C, Wang ZQ, Qiu MZ, Luo HY, Zhang DS, Wang FH, Li YH, Xu RH. ABO blood group, hepatitis B viral infection and risk of pancreatic cancer. Int J Cancer 2011; 131:461-8. [PMID: 21858814 DOI: 10.1002/ijc.26376] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 08/10/2011] [Indexed: 12/14/2022]
Abstract
Little is known about the role of association between ABO blood type and risk of pancreatic cancer develops through effects on hepatitis B viral (HBV) infection. Our study aimed to determine whether joint ABO blood type and HBV infection could increase the risk for pancreatic cancer. A total of 645 patients with pancreatic adenocarcinoma and 711 age- and sex-matched individuals who had nonmalignant diseases treated at the Sun Yat-sen University Cancer Center in China were retrospectively analyzed. Blood samples were tested for ABO blood type and hepatitis B surface antigen (HBsAg), hepatitis B surface antibody (anti-HBs), hepatitis B e antigen (HBeAg), hepatitis B e antibody (anti-HBe) and hepatitis B core antibody (anti-HBc). Multivariable unconditional logistic regression analysis was used to estimate adjusted odds ratios [AORs] and 95% confidence interval [CI]. Multivariable analysis with adjustment for risk factors showed that A blood type, HBsAg-positive/anti-HBc-positive, anti-HBs-positive/anti-HBc-positive were significantly associated with pancreatic cancer. The estimated AORs (95% CI) were as follows: A blood type, 1.425 (1.071-1.894), HBsAg-positive/anti-HBc-positive, 1.610 (1.125-2.304), anti-HBs-positive/anti-HBc-positive, 1.526 (1.159-2.011). The effect of A blood type significantly modified the risk of pancreatic cancer among subjects with anti-HBc-positive (AORs = 1.882, 95% CI, 1.284-2.760). In our study, we reported an association between A blood type, infection with HBV and pancreatic cancer risk. Moreover, we found a synergism between A blood type and HBV infection in the development of pancreatic cancer.
Collapse
Affiliation(s)
- De-Shen Wang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Familial pancreatic cancer and hereditary syndromes: screening strategy for high-risk individuals. J Gastroenterol 2011; 46:1249-59. [PMID: 21847571 DOI: 10.1007/s00535-011-0457-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 07/25/2011] [Indexed: 02/04/2023]
Abstract
Globally, and almost evenly across nations, a familial disposition can be found in 4-10% of patients with pancreatic cancer (PC). A family history of PC is a risk for this disease and the risk level changes in correlation with the number of affected relatives. Several hereditary syndromes with potential germline mutation also have a high risk for PC; however, little is yet known regarding the genes responsible for familial pancreatic cancer (FPC). Characteristics of FPC cases are similar to those of other familial tumors, including younger onset than in sporadic cases and an ethnic difference (Ashkenazi Jewish > other Caucasian). Other risks resemble those of sporadic cases and include smoking and diabetes mellitus. People with several genetic syndromes, including Peutz-Jeghers syndrome, hereditary pancreatitis, breast-ovarian cancer syndrome, hereditary nonpolyposis colorectal cancer, and familial adenomatous polyposis also have an increased risk of PC. In many countries, but not yet in Japan, screening of these high-risk individuals is now ongoing for the detection of early PC under established familial pancreatic cancer registries. In addition to the ordinary risk factors, such as smoking, diabetes, pancreatitis, cysts, duct ectasia, and intraductal papillary mucinous neoplasm (IPMN), individuals with a family history of PC and hereditary syndromes are expected to be entered into the screening protocol.
Collapse
|
39
|
Abstract
OBJECTIVES In western countries, 7% to 10% of patients with pancreatic cancer (PC) have a familial predisposition to their disease. The aim of this study was to determine the familial susceptibility to PC in Japan. METHODS Five hundred seventy-seven patients with PC and 577 age- and gender-matched controls were analyzed for cancer history in their first-degree relative(s) (FDRs) and demographic factors. RESULTS The patients with PC were more likely to have an FDR with PC (6.9%) than the controls (2.9%; odds ratio [OR], 2.5; P = 0.02). Three patients (0.5%), but none of the controls, had a family history of PC in multiple FDRs. Smoking, especially current smoking (OR, 1.5; P = 0.005), and diabetes mellitus (OR: 1.7, P = 0.001) were also associated with PC. The odds increased up to 10-fold if the patients were positive for these 3 factors. The patients with familial PC were more likely to be current smokers (40%) and to have diabetes mellitus (32.5%) than the sporadic cases (30.1% and 20.1%; OR, 1.6 and 1.9). CONCLUSIONS A family history of PC is a risk of PC in Japan (6.9%) as is a personal history of diabetes and smoking. It is prudent to inform the kindred of patients with familiar PC of the risk of smoking and to follow carefully if they develop diabetes.
Collapse
|
40
|
Screening for pancreatic cancer in a high-risk population with serum CA 19-9 and targeted EUS: a feasibility study. Gastrointest Endosc 2011; 74:87-95. [PMID: 21704809 DOI: 10.1016/j.gie.2011.03.1235] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 03/21/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND Earlier detection of pancreatic adenocarcinoma is needed. OBJECTIVE To determine whether early pancreatic neoplasia can be detected in a high-risk population by using CA 19-9 followed by targeted EUS. DESIGN Prospective cohort study. SETTING Two academic medical centers. PATIENTS Eligible patients met age criteria and had at least 1 first-degree relative with pancreatic adenocarcinoma. INTERVENTIONS A serum CA 19-9 was performed on all patients. EUS was performed if the CA 19-9 level was elevated. FNA of identified lesions was performed. Patients with pancreatic cancer detected by using this screening protocol were compared with patients presenting off-protocol for staging data. Medicare reimbursement rates were used to derive cost data. MAIN OUTCOME MEASUREMENTS Detection of early pancreatic neoplasia. RESULTS A total of 546 patients were enrolled. CA 19-9 was elevated in 27 patients (4.9%, 95% CI, 3.2%-7.1%). Neoplastic or malignant findings were detected in 5 patients (0.9%, 95% CI, 0.3%-2.1%), and pancreatic adenocarcinoma in 1 patient (0.2%, 95% CI, 0.005%-1.02%). The patient with pancreatic cancer detected as part of this protocol was 1 of 2 patients presenting to the University of Vermont with stage 1 cancer. The cost to detect 1 pancreatic neoplasia was $8431. The cost to detect 1 pancreatic adenocarcinoma was $41,133. LIMITATIONS The sample size is adequate only to demonstrate the feasibility of this approach. CONCLUSIONS Potentially curative pancreatic adenocarcinoma can be identified with this screening protocol. Stage 1 pancreatic cancer is more likely to be detected by using this screening protocol than by using standard means of detection.
Collapse
|
41
|
Family history of cancer and tobacco exposure in index cases of pancreatic ductal adenocarcinoma. JOURNAL OF ONCOLOGY 2011; 2011:215985. [PMID: 21547248 PMCID: PMC3085295 DOI: 10.1155/2011/215985] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Accepted: 02/09/2011] [Indexed: 12/19/2022]
Abstract
Aim. To examine interaction between history of cancer in first-degree relatives and tobacco smoking in index patients of pancreatic adenocarcinoma.
Methods. We carried out a case-control involving 113 patients with pancreatic adenocarcinoma and 110 controls over a 12-month period at the Freeman Hospital, Newcastle upon Tyne, UK. They were all administered a detailed tobacco exposure questionnaire and a family history questionnaire. We calculated cumulative tobacco exposure and risk for pancreas cancer.
Results. Both smokers (OR 3.01 (95% CI: 1.73 to 5.24)) and those with a family history of malignancy (OR 1.98 (95% CI: 1.15–3.38)) were more likely to develop pancreatic cancer. Having more than one first-degree relative with cancer did not significantly further increase the risk of pancreatic cancer. Amongst pancreatic cancer cases, cumulative tobacco exposure was significantly decreased (P = .032) in the group of smokers (current and ex-smokers) who had a family history of malignancy [mean (SD): 30.00 (24.77) pack-years versus 44.69 (28.47) pack-years with no such history].
Conclusions. Individuals with a family history of malignancy are at an increased risk of pancreatic cancer. Furthermore, individuals with a family history of malignancy and who smoke appear to require a lesser degree of tobacco exposure for the development of pancreatic cancer.
Collapse
|
42
|
Ward CJ, Wu Y, Johnson RA, Woollard JR, Bergstralh EJ, Cicek MS, Bakeberg J, Rossetti S, Heyer CM, Petersen GM, Lindor NM, Thibodeau SN, Harris PC, Torres VE, Hogan MC, Boardman LA. Germline PKHD1 mutations are protective against colorectal cancer. Hum Genet 2011; 129:345-9. [PMID: 21274727 DOI: 10.1007/s00439-011-0950-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/12/2011] [Indexed: 12/19/2022]
Abstract
The autosomal recessive polycystic kidney disease (ARPKD) gene, PKHD1, has been implicated in the genesis or growth of colorectal adenocarcinoma, as a high level of somatic mutations was found in colorectal tumor tissue. To determine whether carriers of a single PKHD1 mutation are at increased risk of colorectal carcinoma, we assessed the prevalence of the commonest European mutation, T36M. First, we assayed a European cohort of ARPKD patients and found T36M was responsible for 13.1% of mutations. We then investigated two European cohorts with colorectal adenocarcinoma versus two control cohorts of similar age and gender. Screening for the most common PKHD1 mutation, T36M, we detected 15:3,603 (0.42%) controls versus 1:3,767 (0.027%) colorectal cancer individuals, indicating that heterozygous PKHD1 mutations are not a risk factor and are protective (p=0.0002). We also show that the carriage rate for PKHD1 mutations in the European population is higher than previous accepted at 3.2% (1:31 genomes).
Collapse
Affiliation(s)
- Christopher J Ward
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Affiliation(s)
- Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, 401 North Broadway, Weinberg 2242, Baltimore, MD 21231, USA.
| | | | | | | | | |
Collapse
|
44
|
Jacobs EJ, Chanock SJ, Fuchs CS, Lacroix A, McWilliams RR, Steplowski E, Stolzenberg-Solomon RZ, Arslan AA, Bueno-de-Mesquita HB, Gross M, Helzlsouer K, Petersen G, Zheng W, Agalliu I, Allen NE, Amundadottir L, Boutron-Ruault MC, Buring JE, Canzian F, Clipp S, Dorronsoro M, Gaziano JM, Giovannucci EL, Hankinson SE, Hartge P, Hoover RN, Hunter DJ, Jacobs KB, Jenab M, Kraft P, Kooperberg C, Lynch SM, Sund M, Mendelsohn JB, Mouw T, Newton CC, Overvad K, Palli D, Peeters PHM, Rajkovic A, Shu XO, Thomas G, Tobias GS, Trichopoulos D, Virtamo J, Wactawski-Wende J, Wolpin BM, Yu K, Zeleniuch-Jacquotte A. Family history of cancer and risk of pancreatic cancer: a pooled analysis from the Pancreatic Cancer Cohort Consortium (PanScan). Int J Cancer 2010; 127:1421-8. [PMID: 20049842 PMCID: PMC2926939 DOI: 10.1002/ijc.25148] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A family history of pancreatic cancer has consistently been associated with increased risk of pancreatic cancer. However, uncertainty remains about the strength of this association. Results from previous studies suggest a family history of select cancers (i.e., ovarian, breast and colorectal) could also be associated, although not as strongly, with increased risk of pancreatic cancer. We examined the association between a family history of 5 types of cancer (pancreas, prostate, ovarian, breast and colorectal) and risk of pancreatic cancer using data from a collaborative nested case-control study conducted by the Pancreatic Cancer Cohort Consortium. Cases and controls were from cohort studies from the United States, Europe and China, and a case-control study from the Mayo Clinic. Analyses of family history of pancreatic cancer included 1,183 cases and 1,205 controls. A family history of pancreatic cancer in a parent, sibling or child was associated with increased risk of pancreatic cancer [multivariate-adjusted odds ratios (ORs) = 1.76, 95% confidence interval (CI) = 1.19-2.61]. A family history of prostate cancer was also associated with increased risk (OR = 1.45, 95% CI = 1.12-1.89). There were no statistically significant associations with a family history of ovarian cancer (OR = 0.82, 95% CI = 0.52-1.31), breast cancer (OR = 1.21, 95% CI = 0.97-1.51) or colorectal cancer (OR = 1.17, 95% CI = 0.93-1.47). Our results confirm a moderate sized association between a family history of pancreatic cancer and risk of pancreatic cancer and also provide evidence for an association with a family history of prostate cancer worth further study.
Collapse
Affiliation(s)
- Eric J Jacobs
- Department of Epidemiology, American Cancer Society, Atlanta, GA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Shirts BH, Burt RW, Mulvihill SJ, Cannon-Albright LA. A population-based description of familial clustering of pancreatic cancer. Clin Gastroenterol Hepatol 2010; 8:812-6. [PMID: 20570637 DOI: 10.1016/j.cgh.2010.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/04/2010] [Accepted: 05/06/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Several familial pancreatic cancer syndromes have been identified. However, the prevalence of familial pancreatic cancers in the general population has not been well defined. METHODS We linked pancreatic cancer cases, identified through the Utah Cancer Registry, to the Utah Population Database, which contains genealogic data from Utah pioneers and their descendants. This database includes 1411 pancreatic adenocarcinoma cases with 3 or more generations of Utah pioneer genealogy. We examined the familial clustering of pancreatic cancer by evaluating the relative risk (RR) of pancreatic cancer among relatives of cases. We also used the genealogical index of familiality to test the hypothesis of no excess relatedness among pancreatic cancer cases. RESULTS The risk of pancreatic cancer was significantly increased in first-degree (RR, 1.84; 95% confidence interval [CI], 1.47-2.29; P < .0001) and second-degree (RR, 1.59; 95% CI, 1.31-2.91; P < .0001) relatives of individuals with pancreatic cancer. Analysis of case relatedness indicated significant excess relatedness for pancreatic cancer. More than 300 high-risk pedigrees were identified, with from 3-14 cases observed among descendants of pedigree founders. CONCLUSIONS This population-based study provides evidence for increased risk of pancreatic cancer among relatives of cases and for a significantly higher average relatedness among cases than expected. These observations support the role of genetic factors in pancreatic cancer.
Collapse
Affiliation(s)
- Brian H Shirts
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | | | | | | |
Collapse
|
46
|
Genome-wide association study of pancreatic cancer in Japanese population. PLoS One 2010; 5:e11824. [PMID: 20686608 PMCID: PMC2912284 DOI: 10.1371/journal.pone.0011824] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Accepted: 07/03/2010] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer shows very poor prognosis and is the fifth leading cause of cancer death in Japan. Previous studies indicated some genetic factors contributing to the development and progression of pancreatic cancer; however, there are limited reports for common genetic variants to be associated with this disease, especially in the Asian population. We have conducted a genome-wide association study (GWAS) using 991 invasive pancreatic ductal adenocarcinoma cases and 5,209 controls, and identified three loci showing significant association (P-value<5×10−7) with susceptibility to pancreatic cancer. The SNPs that showed significant association carried estimated odds ratios of 1.29, 1.32, and 3.73 with 95% confidence intervals of 1.17–1.43, 1.19–1.47, and 2.24–6.21; P-value of 3.30×10−7, 3.30×10−7, and 4.41×10−7; located on chromosomes 6p25.3, 12p11.21 and 7q36.2, respectively. These associated SNPs are located within linkage disequilibrium blocks containing genes that have been implicated some roles in the oncogenesis of pancreatic cancer.
Collapse
|
47
|
Mai PL, Wideroff L, Greene MH, Graubard BI. Prevalence of family history of breast, colorectal, prostate, and lung cancer in a population-based study. Public Health Genomics 2010; 13:495-503. [PMID: 20389042 DOI: 10.1159/000294469] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 11/17/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND A positive family history is a known risk factor for several cancers; thus, obtaining a thorough family cancer history is essential in cancer risk evaluation and prevention management. METHODS The Family Health Study, a telephone survey in Connecticut, was conducted in 2001. A total of 1,019 participants with demographic information and family cancer history were included in this study. Prevalence of a positive family history of breast, colorectal, prostate, and lung cancer for first- and second-degree relatives was estimated. Logistic regression was used to compare prevalence by demographic factors. RESULTS A positive family history among first-degree relatives was reported by 10.9% (95% Confidence Interval, CI = 8.8-13.3) of respondents for breast cancer, 5.1% (95% CI = 3.9-6.7) for colorectal cancer, 7.0% (95% CI = 5.2-9.4) for prostate cancer, and 6.4% (95% CI = 4.9-8.3) for lung cancer. The reported prevalence of family history of specific cancers varied by sex, age and race/ethnicity of the respondents. CONCLUSION Family history prevalence for 4 of the most common adult solid tumors is substantial and the reported prevalence varied by respondent characteristics. Additional studies are needed to evaluate tools to promote accurate reporting of family history of cancer.
Collapse
Affiliation(s)
- P L Mai
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20852, USA.
| | | | | | | |
Collapse
|
48
|
Naccarati A, Pardini B, Polakova V, Smerhovsky Z, Vodickova L, Soucek P, Vrana D, Holcatova I, Ryska M, Vodicka P. Genotype and haplotype analysis of TP53 gene and the risk of pancreatic cancer: an association study in the Czech Republic. Carcinogenesis 2010; 31:666-70. [PMID: 20110284 DOI: 10.1093/carcin/bgq032] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pancreatic carcinoma is the fourth leading cause of cancer-related deaths in the Czech Republic, with only a minimum of patients surviving 5 years. The aetiology and molecular pathogenesis are still weakly understood. TP53 has a fundamental role in cell cycle and apoptosis and is frequently mutated in solid tumours, including pancreatic cancer. Based on the assumption that genetic variation may affect susceptibility to cancer development, the role of TP53 polymorphisms in modulating the risk of pancreatic cancer may be of major importance. We investigated four selected polymorphisms in TP53 (rs17878362:A(1)>A(2), rs1042522:G>C, rs12947788:C>T and rs17884306:G>A) in association with pancreatic cancer risk in a case-control study, including 240 cases and controls (for a total of 1827 individuals) from the Czech Republic. Carriers of the variant C allele of rs1042522 polymorphism were at an increased risk of pancreatic cancer [odds ratio (OR) 1.73; 95% confidence interval (CI) 1.26-2.39; P = 0.001]. Haplotype analysis showed that in comparison with the most common haplotype (A(1)GCG), the A(2)CCG haplotype was associated with an increased risk (OR 1.39; 95% CI 1.02-1.88; P = 0.034) and the A(1)CCG with a reduced risk (OR 0.30; 95% CI 0.12-0.76; P = 0.011) for this cancer. These results reflect previous findings of a recent association study, where haplotypes constructed on the same TP53 variants were associated with colorectal cancer risk [Polakova et al. (2009) Genotype and haplotype analysis of cell cycle genes in sporadic colorectal cancer in the Czech Republic. Hum. Mutat., 30, 661-668.]. Genetic variation in TP53 may contribute, alone or in concert with other risk factors, to modify the inherited susceptibility to pancreatic cancer, as well as to other gastrointestinal cancers.
Collapse
Affiliation(s)
- A Naccarati
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Academy of Sciences of Czech Republic, Videnska 1083, 14200 Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
López Serrano A. [Risk factors and early diagnosis of pancreatic cancer]. GASTROENTEROLOGIA Y HEPATOLOGIA 2009; 33:382-90. [PMID: 20005016 DOI: 10.1016/j.gastrohep.2009.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 10/20/2009] [Indexed: 12/18/2022]
Abstract
Pancreatic cancer (PC) is usually incurable. Identifying people at risk for developing PC may improve the prognosis of this entity. The main risk factors for PC are Peutz-Jeghers syndrome, hereditary pancreatitis and a history family of PC. Other factors, such as advanced age and smoking, should also be taken into account. PC screening is only useful in very high risk individuals. Tools that allow tumors to be identified in the early stages are required in order to apply appropriate curative treatments. In this scenario, only endoscopic ultrasound with cytological analysis of suspicious pancreatic lesions has proved to be useful.
Collapse
Affiliation(s)
- Antonio López Serrano
- Servicio de Medicina Digestiva, Hospital Universitario Manises, Universidad Católica de Valencia, Valencia, España.
| |
Collapse
|
50
|
Hiripi E, Lorenzo Bermejo J, Li X, Sundquist J, Hemminki K. Familial association of pancreatic cancer with other malignancies in Swedish families. Br J Cancer 2009; 101:1792-7. [PMID: 19826425 PMCID: PMC2778532 DOI: 10.1038/sj.bjc.6605363] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background: The aim of this study was to characterise the familial association of pancreatic cancer with other malignancies. Methods: Relative risks (RRs) of pancreatic cancer according to family history of cancer were calculated using the updated Swedish Family-Cancer Database, which includes over 11.5 million individuals. Estimates were based on Poisson regression. RRs of tumours for individuals with a parental history of pancreatic cancer were also estimated. Results: The risk of pancreatic cancer was elevated in individuals with a parental history of cancers of the liver (RR 1.41; 95% CI 1.10–1.81), kidney (RR 1.37; 95% CI 1.06–1.76), lung (RR 1.50; 95% CI 1.27–1.79) and larynx (RR 1.98; 95% CI 1.19–3.28). Associations were also found between parental history of pancreatic cancer and cancers of the small intestine, colon, breast, lung, testis and cervix in offspring. There was an increased risk of pancreatic cancer associated with early-onset breast cancer in siblings. Conclusion: Pancreatic cancer aggregates in families with several types of cancer. Smoking may contribute to the familial aggregation of pancreatic and lung tumours, and the familial clustering of pancreatic and breast cancer could be partially explained by inherited mutations in the BRCA2 gene.
Collapse
Affiliation(s)
- E Hiripi
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ) Im Neuenheimer Feld 580, 69120, Germany.
| | | | | | | | | |
Collapse
|