1
|
Mohamed G, Munir M, Rai A, Gaddam S. Pancreatic Cancer: Screening and Early Detection. Gastroenterol Clin North Am 2025; 54:205-221. [PMID: 39880528 DOI: 10.1016/j.gtc.2024.09.006] [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] [Indexed: 01/31/2025]
Abstract
Pancreatic cancer, often diagnosed at advanced stages, has poor survival rates. Effective screening aims to detect the disease early, improving outcomes. Current guidelines recommend screening high-risk groups, including those with a family history or genetic predispositions, using methods like endoscopic ultrasound and MRI. The American Gastroenterological Association and other organizations advise annual surveillance for high-risk individuals, typically starting at the age of 50 or 10 years younger than the youngest affected relative. For certain genetic syndromes, such as Peutz-Jeghers syndrome or hereditary pancreatitis, screening may begin as early as the age of 35 to 40 years.
Collapse
Affiliation(s)
- Ghada Mohamed
- Department of Internal Medicine, Lahey Hospital & Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Malak Munir
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Amar Rai
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA
| | - Srinivas Gaddam
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, ST, Suite 7705, Los Angeles, CA 90048, USA.
| |
Collapse
|
2
|
Luo D, Li Y, Yu X, Ji L, Gong X. Early onset pancreatic cancer: A review. Transl Oncol 2025; 52:102239. [PMID: 39672003 PMCID: PMC11699111 DOI: 10.1016/j.tranon.2024.102239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/17/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024] Open
Abstract
Early-onset pancreatic cancer (EOPC) is usually defined as patients with pancreatic cancer before the age of 50 years, which is relatively rare. However, the research on EOPC is somewhat obscure, and the specific clinical and molecular characteristics of this condition are debated. In this review, we discussed the differences between EOPC and late-onset pancreatic cancer (LOPC) or average-onset pancreatic cancer (AOPC) with a focus on clinical and molecular characteristics, survival outcomes and treatment to promote the diagnosis and treatment of EOPC.
Collapse
Affiliation(s)
- Dong Luo
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Department of General, Visceral and Transplant Surgery, European Pancreas Centre, Heidelberg. University Hospital, Heidelberg, Germany
| | - Yixiong Li
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery Ⅱ, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Liandong Ji
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| | - Xuejun Gong
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
3
|
Brentnall TA. Familial pancreatic cancer: a long fruitful journey. Fam Cancer 2024; 23:217-220. [PMID: 38436765 DOI: 10.1007/s10689-024-00364-5] [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: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 03/05/2024]
Abstract
In the early years of my GI fellowship, a healthy 40-year-old man came to my clinic and announced that he was going to die of pancreatic cancer. His brothers, father and uncles had all died of the disease; he felt his fate was inescapable. I asked whether his family members had seen doctors or had any tests. His answer was yes to both. Even so, doctors could not diagnose the pancreatic cancer at early stages. CT scans were always negative. I thought to myself, in order to help this patient-CT scans may not be reliable for early detection. Perhaps other methods of imaging the pancreas might be of more benefit. This patient opened a door that led to a 30-year journey of trying to detect pancreatic cancer at earlier stages when it is curable.
Collapse
Affiliation(s)
- Teresa A Brentnall
- Department of Medicine, University of Washington, PO Box 356424, 1959 NE Pacific, Seattle, WA, USA.
| |
Collapse
|
4
|
Chandana SR, Woods LM, Maxwell F, Gandolfo R, Bekaii-Saab T. Risk factors for early-onset pancreatic ductal adenocarcinoma: A systematic literature review. Eur J Cancer 2024; 198:113471. [PMID: 38154392 DOI: 10.1016/j.ejca.2023.113471] [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: 09/11/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Emerging cancer trends suggest an increase in pancreatic cancer incidence in individuals younger than its typical age of onset, potentially reflecting changes in population exposures and lifestyles. PATIENTS AND METHODS We conducted a PRISMA-standard systematic literature review to identify non-heritable risk factors for early-onset pancreatic ductal adenocarcinoma (PDAC) (PROSPERO number: CRD42022299397). Systematic searches of MEDLINE and Embase bibliographic databases were performed (January 2022), and publications were screened against predetermined eligibility criteria; data were extracted using standardised data fields. The STROBE checklist was used to assess the completeness of reporting as a proxy for publication quality. Data were categorised by risk factor and analysed descriptively. RESULTS In total, 24 publications were included. All publications reported observational study data; thresholds for age group comparisons ranged between 40 and 65 years. Lifestyle factors investigated included smoking, alcohol consumption, obesity, physical inactivity, meat intake, socioeconomic status and geographical residence. Clinical factors investigated included pancreatitis, diabetes/insulin resistance, prior cancer and cancer stage at diagnosis, hepatitis B infection, metabolic syndrome and long-term proton pump inhibitor exposure. Publication STROBE scores were 6-21 (maximum, 22). Eight studies reported results adjusted for confounders. Potential non-heritable risk factors for early-onset PDAC that warrant further investigation included smoking, alcohol consumption, pancreatitis and hepatitis B infection. CONCLUSION Evidence for non-heritable risk factors for early-onset PDAC is heterogeneous, but four factors were identified that might aid the identification of at-risk individuals who may benefit from screening and risk reduction strategies.
Collapse
Affiliation(s)
- Sreenivasa R Chandana
- Department of Gastrointestinal Medical Oncology, The Cancer and Hematology Centers, Grand Rapids, MI, USA.
| | - Laura M Woods
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | | | | | | |
Collapse
|
5
|
Genc D, Ozbek O, Oral B, Yıldırım R, Ileri Ercan N. Phytochemicals in Pancreatic Cancer Treatment: A Machine Learning Study. ACS OMEGA 2024; 9:413-421. [PMID: 38222639 PMCID: PMC10785644 DOI: 10.1021/acsomega.3c05861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/17/2023] [Accepted: 11/28/2023] [Indexed: 01/16/2024]
Abstract
The discovery of new strategies and novel therapeutic agents is crucial to improving the current treatment methods and increasing the efficacy of cancer therapy. Phytochemicals, naturally occurring bioactive constituents derived from plants, have great potential in preventing and treating various diseases, including cancer. This study reviewed 74 literature studies published between 2006 and 2022 that conducted in vitro cytotoxicity and cell apoptosis analyses of the different concentrations of phytochemicals and their combinations with conventional drugs or supplementary phytochemicals on human pancreatic cell lines. From 34 plant-derived phytochemicals on 20 human pancreatic cancer cell lines, a total of 11 input and 2 output variables have been used to construct the data set that contained 2161 different instances. The machine learning approach has been implemented using random forest for regression, whereas association rule mining has been used to determine the effects of individual phytochemicals. The random forest models developed are generally good, indicating that the phytochemical type, its concentration, and the type of cell line are the most important descriptors for predicting the cell viability. However, for predicting cell apoptosis the primary phytochemical type is the most significant descriptor . Among the studied phytochemicals, catechin and indole-3-carbinol were found to be non-cytotoxic at all concentrations irrespective of the treatment time. On the other hand, berbamine and resveratrol were strongly cytotoxic with cell viabilities of less than 40% at a concentration range between 10 and 100 μM and above 100 μM, respectively, which brings them forward as potential therapeutic agents in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Destina
Ekingen Genc
- Department
of Chemical Engineering, Bogazici University, Bebek, Istanbul 34342, Turkey
| | - Ozlem Ozbek
- Department
of Chemical Engineering, Bogazici University, Bebek, Istanbul 34342, Turkey
| | - Burcu Oral
- Department
of Chemical Engineering, Bogazici University, Bebek, Istanbul 34342, Turkey
| | - Ramazan Yıldırım
- Department
of Chemical Engineering, Bogazici University, Bebek, Istanbul 34342, Turkey
| | - Nazar Ileri Ercan
- Department
of Chemical Engineering, Middle East Technical
University, Çankaya, Ankara 06800, Turkey
| |
Collapse
|
6
|
Seifert L, Weitz J. [Familial pancreatic cancer syndrome]. CHIRURGIE (HEIDELBERG, GERMANY) 2023; 94:406-411. [PMID: 36799964 DOI: 10.1007/s00104-023-01819-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 02/18/2023]
Abstract
In contrast to many other malignant entities the risk factors for pancreatic cancer are mostly unknown. Hereditary factors are causative in about 3-6% of all pancreatic cancers. In addition to hereditary tumor disposition syndromes and hereditary pancreatitis, the familial pancreatic cancer syndrome accounts for about 75% of all hereditary pancreatic cancers. A familial pancreatic cancer syndrome is present when at least two first-degree relatives in a family have histologically proven pancreatic cancer and there are no criteria for another hereditary syndrome. An underlying uniform monogenetic gene defect in familial pancreatic cancer syndrome is currently unknown and a germline mutation in the BRCA2 gene is so far the most frequently identified genetic alteration. Patients at risk for familial pancreatic cancer should be offered participation in registry studies with associated early detection programs (e.g., the FaPaCa registry). These enable the detection of high-grade neoplasms and pancreatic cancer in the early stages, which appears to reduce mortality; however, the detected pancreatic lesions are often incorrectly diagnostically classified, and patients undergo complex pancreatic resection despite benign findings. Finally, studies have so far not been able to clearly clarify whether patients at risk benefit from an early detection program. According to current data, prophylactic operations are not indicated for familial pancreatic cancer syndrome and should only be performed if there is evidence of a suspicious finding that requires resection.
Collapse
Affiliation(s)
- Lena Seifert
- Klinik und Poliklinik für Viszeral‑, Thorax- und Gefäßchirurgie, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| | - Jürgen Weitz
- Klinik und Poliklinik für Viszeral‑, Thorax- und Gefäßchirurgie, Universitätsklinikum Carl Gustav Carus an der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| |
Collapse
|
7
|
Druk IV. Pancreatic cancer, pancreatogenic diabetes, type 2 diabetes mellitus. EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2022:171-182. [DOI: 10.31146/1682-8658-ecg-205-9-171-182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Pancreatic cancer (PC) is the fourth leading cause of death among all types of cancer. PC is very aggressive with a low 5-year overall survival rate. The highest prevalence of diabetes mellitus (DM), significantly exceeding the average population, is registered among patients with prostate cancer Recommendations for systemic screening of patients with diabetes for the detection of PC are not standardized. The purpose of this review is to present an analysis of current literature data on pathogenetic relationships between DM and PC and prospects for PC screening. Research data indicate that there is a bidirectional relationship between DM and PC, in which DM can act either as a risk factor for PC or as a marker of paraneoplastic syndrome of PC. In the differential diagnosis of type 2 diabetes, pancreatogenic diabetes and diabetes associated with PC, a set of clinical signs can be used. Patients with DM who have additional signs/symptoms of increased risk can be considered as a group subject to mandatory screening. Numerous studies of various proteomic, metabolomic, genetic and transcriptomic biomarkers PC have been published. The search for an easy-to-use clinically useful and cost-effective PC marker is still ongoing.
Collapse
|
8
|
Klatte DCF, Wallace MB, Löhr M, Bruno MJ, van Leerdam ME. Hereditary pancreatic cancer. Best Pract Res Clin Gastroenterol 2022; 58-59:101783. [PMID: 35988957 DOI: 10.1016/j.bpg.2021.101783] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/09/2021] [Accepted: 12/15/2021] [Indexed: 01/31/2023]
Abstract
Pancreatic cancer is one of the deadliest malignancies. Therefore, there is an urgent need to detect pancreatic cancer in an earlier stage to improve outcomes. A variety of hereditary cancer syndromes have been associated with an increased risk of developing pancreatic cancer, and these individuals may benefit from surveillance programs. Surveillance programs have shown potential to improve outcomes, but have important risks such as overtreatment. In this review we will discuss the definitions and epidemiology of hereditary pancreatic cancer, recommendations for genetic testing and participation in surveillance. Important aspects are differences in surveillance strategies, target lesions, and potential benefits and harms of surveillance. Lastly we will highlight future directions for research and improvement of care for individuals at high-risk of pancreatic cancer.
Collapse
Affiliation(s)
- Derk C F Klatte
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, United States.
| | - Michael B Wallace
- Department of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, United States; Division of Gastroenterology and Hepatology, Sheikh Shakhbout Medical City, Abu Dhabi, United Arab Emirates.
| | - Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| | - Marco J Bruno
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, the Netherlands.
| | - Monique E van Leerdam
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands; Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Lin RT, Chen PL, Yang CY, Yeh CC, Lin CC, Huang WH, Chung AK, Lin JT. Risk factors related to age at diagnosis of pancreatic cancer: a retrospective cohort pilot study. BMC Gastroenterol 2022; 22:243. [PMID: 35568803 PMCID: PMC9107247 DOI: 10.1186/s12876-022-02325-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Increased pancreatic cancer incidence has been observed among younger than in older adults. This pilot study aimed to determine the feasibility of a large study that would compare the age at diagnosis of pancreatic cancer among patients with different risk factors. METHODS We compared the age at diagnosis of pancreatic cancer between groups of pancreatic cancer patients exposed and not exposed to the identified risk factors. We estimated the age at which exposure started, average exposure quantity, and total years of exposure and investigated their relationships with age at diagnosis of pancreatic cancer. RESULTS Sixteen out of 24 (67%) subjects carried known genetic factors and/or had smoking and/or drinking habits; however, an earlier age of pancreatic cancer diagnosis was not observed. Conversely, we found a significant correlation between the age at which alcohol consumption was started and the age at diagnosis of pancreatic cancer (r = 0.8124, P = 0.0043). CONCLUSIONS Our pilot study suggested that a large study following this study design is feasible and that the following should be conducted in a large study: mediation analysis for disease-related factors, advanced genomic analysis for new candidate genes, and the correlation between age of first exposure to risk factors and pancreatic cancer onset.
Collapse
Affiliation(s)
- Ro-Ting Lin
- Department of Occupational Safety and Health, College of Public Health, China Medical University, Taichung, 406040 Taiwan
| | - Pei-Lung Chen
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, 100226 Taiwan
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100025 Taiwan
| | - Chi-Ying Yang
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, 404332 Taiwan
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung, 404332 Taiwan
- Department of Surgery, Asia University Hospital, Taichung, 413505 Taiwan
| | - Chun-Che Lin
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, 404332 Taiwan
| | - Wen-Hsin Huang
- Department of Internal Medicine, Digestive Medicine Center, China Medical University Hospital, Taichung, 404332 Taiwan
| | - An-Ko Chung
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100025 Taiwan
| | - Jaw-Town Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, No. 1, Yida Rd., Yanchao Dist., Kaohsiung, 824005 Taiwan
| |
Collapse
|
10
|
Pauley K, Khan A, Kohlmann W, Jeter J. Considerations for Germline Testing in Melanoma: Updates in Behavioral Change and Pancreatic Surveillance for Carriers of CDKN2A Pathogenic Variants. Front Oncol 2022; 12:837057. [PMID: 35372037 PMCID: PMC8967159 DOI: 10.3389/fonc.2022.837057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/23/2022] [Indexed: 01/13/2023] Open
Abstract
The largest proportion of hereditary melanoma cases are due to pathogenic variants (PVs) in the CDKN2A/p16 gene, which account for 20%-40% of familial melanomas and confer up to a 30%-70% lifetime risk for melanoma in individuals with these variants. In addition, PVs in the CDKN2A gene also increase risk for pancreatic cancer (~5-24% lifetime risk). Individuals with PVs in the CDKN2A gene also tend to have an earlier onset of cancer. Despite these known risks, uptake of germline testing has been limited in the past, largely due to perceptions of limited benefit for patients. Prevention recommendations have been developed for individuals with CDKN2A PVs as well the providers who care for them. On the patient level, behavioral modifications regarding melanoma prevention such as wearing sunscreen, limiting prolonged sun exposure and practicing general sun safety can help reduce risks. Germline testing can provide motivation for some individuals to adhere to these lifestyle changes. On the provider level, pancreatic cancer surveillance for individuals with CDKN2A PVs has been increasingly endorsed by expert consensus, although the efficacy of these surveillance methods remains under study. This review summarizes the updated surveillance guidelines for individuals with CDKN2A PVs and explores the impact of genetic counseling and testing in influencing behavioral changes in these individuals.
Collapse
Affiliation(s)
- Kristen Pauley
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Ambreen Khan
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Wendy Kohlmann
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Joanne Jeter
- Department of Internal Medicine, Huntsman Cancer Institute, Salt Lake City, UT, United States
| |
Collapse
|
11
|
Antwi SO, Rabe KG, Bamlet WR, Meyer M, Chandra S, Fagan SE, Hu C, Couch FJ, McWilliams RR, Oberg AL, Petersen GM. Influence of Cancer Susceptibility Gene Mutations and ABO Blood Group of Pancreatic Cancer Probands on Concomitant Risk to First-Degree Relatives. Cancer Epidemiol Biomarkers Prev 2022; 31:372-381. [PMID: 34782396 PMCID: PMC8825751 DOI: 10.1158/1055-9965.epi-21-0745] [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: 06/16/2021] [Revised: 09/16/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND ABO blood group is associated with pancreatic cancer risk. Whether ABO blood group alone or when combined with inherited mutation status of index pancreatic cancer cases (probands) can enhance pancreatic cancer risk estimation in first-degree relatives (FDR) is unclear. We examined FDRs' risk for pancreatic cancer based on probands' ABO blood group and probands' cancer susceptibility gene mutation status. METHODS Data on 23,739 FDRs, identified through 3,268 pancreatic cancer probands, were analyzed. Probands' ABO blood groups were determined serologically or genetically, and 20 cancer susceptibility genes were used to classify probands as "mutation-positive" or "mutation-negative." SIRs and 95% confidence intervals (CI) were calculated, comparing observed pancreatic cancer cases in the FDRs with the number expected in SEER-21 (reference population). RESULTS Overall, FDRs had 2-fold risk of pancreatic cancer (SIR = 2.00; 95% CI = 1.79-2.22). Pancreatic cancer risk was higher in FDRs of mutation-positive (SIR = 3.80; 95% CI = 2.81-5.02) than mutation-negative (SIR = 1.79; 95% CI = 1.57-2.04) probands (P < 0.001). The magnitude of risk did not differ by ABO blood group alone (SIRblood-group-O = 1.57; 95% CI = 1.20-2.03, SIRnon-O = 1.83; 95% CI = 1.53-2.17; P = 0.33). Among FDRs of probands with non-O blood group, pancreatic cancer risk was higher in FDRs of mutation-positive (SIR = 3.98; 95% CI = 2.62-5.80) than mutation-negative (SIR = 1.66; 95% CI = 1.35-2.03) probands (P < 0.001), but risk magnitudes were statistically similar when probands had blood group O (SIRmutation-positive = 2.65; 95% CI = 1.09-5.47, SIRmutation-negative = 1.48; 95% CI = 1.06-5.47; P = 0.16). CONCLUSIONS There is a range of pancreatic cancer risk to FDRs according to probands' germline mutation status and ABO blood group, ranging from 1.48 for FDRs of probands with blood group O and mutation-negative to 3.98 for FDRs of probands with non-O blood group and mutation-positive. IMPACT Combined ABO blood group and germline mutation status of probands can inform pancreatic cancer risk estimation in FDRs.
Collapse
Affiliation(s)
- Samuel O Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida.
| | - Kari G Rabe
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - William R Bamlet
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Margaret Meyer
- Department of Medical and Molecular Genetics, Indiana University, Bloomington, Indiana
| | - Shruti Chandra
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Sarah E Fagan
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland
| | - Chunling Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Ann L Oberg
- Division of Computational Biology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Gloria M Petersen
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
12
|
Llach J, Carballal S, Moreira L. Familial Pancreatic Cancer: Current Perspectives. Cancer Manag Res 2020; 12:743-758. [PMID: 32099470 PMCID: PMC6999545 DOI: 10.2147/cmar.s172421] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/15/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer (PC) is a highly lethal disease, mostly incurable when detected. Thus, despite advances in PC treatments, only around 7% of patients survive 5-years after diagnosis. This morbid outcome is secondary to multifactorial reasons, such as late-stage diagnosis, rapid progression and minimal response to chemotherapy. Based on these factors, it is of special relevance to identify PC high-risk individuals in order to establish preventive and early detection measures. Although most PC are sporadic, approximately 10% cases have a familial basis. No main causative gene of PC has been identified but several known germline pathogenic mutations are related with an increased risk of this tumor. These inherited cancer syndromes represent 3% of all PC. On the other hand, in 7% of cases of PC, there is a strong family history without a causative germline mutation, a situation known as familial pancreatic cancer (FPC). In recent years, there is increasing evidence supporting the benefit of genetic germline analysis in PC patients, and periodic pancreatic screening in PC high-risk patients (mainly those with a lifetime risk greater than 5%), although there is no general agreement in the group of patients and individuals to study and screen. In the present review, we expose an update in the field of hereditary and FPC, with the aim of describing the current strategies and implications in genetic counseling, surveillance and therapeutic interventions.
Collapse
Affiliation(s)
- Joan Llach
- Departmento de Gastroenterología, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d' Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| | - Sabela Carballal
- Departmento de Gastroenterología, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d' Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| | - Leticia Moreira
- Departmento de Gastroenterología, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d' Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Wu H, Ding F, Chen X, Chen S, Shi Z, Liu Q, Zheng Z, Chen Y. PRSS1 genotype is associated with prognosis in patients with pancreatic ductal adenocarcinoma. Oncol Lett 2019; 19:121-126. [PMID: 31897122 PMCID: PMC6924091 DOI: 10.3892/ol.2019.11097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/16/2019] [Indexed: 12/21/2022] Open
Abstract
The prognostic value of the genotype of the PRSS1 gene in patients with pancreatic ductal adenocarcinoma (PDAC) remains poorly understood. The aim of the present study was to evaluate the association between the PRSS1 genotype and clinicopathological characteristics of patients with PDAC, as well as to explore the prognostic significance of the PRSS1 genotype in patients with PDAC. A total of 124 patients with PDAC patients were included in the current study and the PRSS1 genotype of the enrolled patients was determined by the polymerase chain reaction. Associations between the PRSS1 genotype and clinicopathological characteristics were subsequently analyzed using the Chi-square test. The impact of the PRSS1 genotype on patient prognosis was assessed using the Kaplan-Meier method, and predictive factors of overall survival (OS) time were analyzed by Cox regression. A total of 56 patients with PDAC (45.16%) had the T/C PRSS1 genotype, which was associated with large tumor sizes (P=0.027) and higher tumor node metastasis (TNM) stages (P=0.041). Following a median follow-up of 19 months, the T/C genotype of PRSS1 genotype was associated with a shorter OS time (P=0.037) compared with the C/C or T/T PRSS1 genotypes. Univariate and multivariate analyses revealed that PRSS1 genotype was identified to be an independent prognostic factor for the OS time of patients with PDAC. The results obtained in the current study suggested that the PRSS1 genotype, as well as factors such as the serum level of carbohydrate antigen 19-9 and the TNM stage, may act as independent prognostic factors for the OS time of patients with PDAC.
Collapse
Affiliation(s)
- Huasheng Wu
- Department of Hepatobiliary Surgery, San Ming First Hospital, Sanming, Fujian 365000, P.R. China
| | - Fadian Ding
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Xijun Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Shaoqin Chen
- Department of Critical Care Medicine, Xiamen Cardiovascular Hospital Xiamen University, Xiamen, Fujian 361000, P.R. China
| | - Zhen Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Qicai Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Zhenhua Zheng
- Department of Hepatobiliary Surgery, San Ming First Hospital, Sanming, Fujian 365000, P.R. China
| | - Youting Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
14
|
Zhang J, Bai R, Li M, Ye H, Wu C, Wang C, Li S, Tan L, Mai D, Li G, Pan L, Zheng Y, Su J, Ye Y, Fu Z, Zheng S, Zuo Z, Liu Z, Zhao Q, Che X, Xie D, Jia W, Zeng MS, Tan W, Chen R, Xu RH, Zheng J, Lin D. Excessive miR-25-3p maturation via N 6-methyladenosine stimulated by cigarette smoke promotes pancreatic cancer progression. Nat Commun 2019; 10:1858. [PMID: 31015415 PMCID: PMC6478927 DOI: 10.1038/s41467-019-09712-x] [Citation(s) in RCA: 280] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/26/2019] [Indexed: 12/15/2022] Open
Abstract
N6-methyladenosine (m6A) modification is an important mechanism in miRNA processing and maturation, but the role of its aberrant regulation in human diseases remained unclear. Here, we demonstrate that oncogenic primary microRNA-25 (miR-25) in pancreatic duct epithelial cells can be excessively maturated by cigarette smoke condensate (CSC) via enhanced m6A modification that is mediated by NF-κB associated protein (NKAP). This modification is catalyzed by overexpressed methyltransferase-like 3 (METTL3) due to hypomethylation of the METTL3 promoter also caused by CSC. Mature miR-25, miR-25-3p, suppresses PH domain leucine-rich repeat protein phosphatase 2 (PHLPP2), resulting in the activation of oncogenic AKT-p70S6K signaling, which provokes malignant phenotypes of pancreatic cancer cells. High levels of miR-25-3p are detected in smokers and in pancreatic cancers tissues that are correlated with poor prognosis of pancreatic cancer patients. These results collectively indicate that cigarette smoke-induced miR-25-3p excessive maturation via m6A modification promotes the development and progression of pancreatic cancer.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Adult
- Aged
- Aged, 80 and over
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/etiology
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor
- Cell Transformation, Neoplastic/genetics
- Co-Repressor Proteins/metabolism
- DNA Methylation
- Disease Progression
- Epithelial Cells/pathology
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- HEK293 Cells
- Humans
- Male
- Methyltransferases/genetics
- Methyltransferases/metabolism
- MicroRNAs/blood
- MicroRNAs/metabolism
- Middle Aged
- Nuclear Proteins/metabolism
- Pancreatic Ducts/cytology
- Pancreatic Ducts/pathology
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/etiology
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Phosphoprotein Phosphatases/genetics
- Phosphoprotein Phosphatases/metabolism
- Prognosis
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA-Binding Proteins/metabolism
- Repressor Proteins
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Smoke/adverse effects
- Smoking/adverse effects
- Smoking/blood
- Nicotiana/toxicity
- Up-Regulation
Collapse
Affiliation(s)
- Jialiang Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ruihong Bai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Huilin Ye
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Wu
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- CAMS Key Laboratory of Genetics and Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengfeng Wang
- Department of Abdominal Surgery, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengping Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liping Tan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Dongmei Mai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guolin Li
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ling Pan
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yanfen Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jiachun Su
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying Ye
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zhiqiang Fu
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shangyou Zheng
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhixiang Zuo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Zexian Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qi Zhao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xu Che
- Department of Abdominal Surgery, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weihua Jia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mu-Sheng Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wen Tan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- CAMS Key Laboratory of Genetics and Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rufu Chen
- Department of Pancreaticobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Rui-Hua Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Jian Zheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Dongxin Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- CAMS Key Laboratory of Genetics and Genomic Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
15
|
Paiella S, Capurso G, Cavestro GM, Butturini G, Pezzilli R, Salvia R, Signoretti M, Crippa S, Carrara S, Frigerio I, Bassi C, Falconi M, Iannicelli E, Giardino A, Mannucci A, Laghi A, Laghi L, Frulloni L, Zerbi A. Results of First-Round of Surveillance in Individuals at High-Risk of Pancreatic Cancer from the AISP (Italian Association for the Study of the Pancreas) Registry. Am J Gastroenterol 2019; 114:665-670. [PMID: 30538291 DOI: 10.1038/s41395-018-0414-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Surveillance programs on high-risk individuals (HRIs) can detect pre-malignant lesions or early pancreatic cancer (PC). We report the results of the first screening round of the Italian multicenter program supported by the Italian Association for the study of the Pancreas (AISP). METHODS The multicenter surveillance program included asymptomatic HRIs with familial (FPC) or genetic frailty (GS: BRCA1/2, p16/CDKN2A, STK11/LKB1or PRSS1, mutated genes) predisposition to PC. The surveillance program included at least an annual magnetic resonance cholangio pancreatography (MRCP). Endoscopic ultrasound (EUS) was proposed to patients who refused or could not be submitted to MRCP. RESULTS One-hundreds eighty-seven HRIs underwent a first-round screening examination with MRCP (174; 93.1%) or EUS (13; 6.9%) from September 2015 to March 2018.The mean age was 51 years (range 21-80).One-hundreds sixty-five (88.2%) FPC and 22 (11.8%) GF HRIs were included. MRCP detected 28 (14.9%) presumed branch-duct intraductal papillary mucinous neoplasms (IPMN), 1 invasive carcinoma/IPMN and one low-grade mixed-type IPMN, respectively. EUS detected 4 PC (2.1%): 1 was resected, 1 was found locally advanced intraoperatively, and 2 were metastatic. Age > 50 (OR 3.3, 95%CI 1.4-8), smoking habit (OR 2.8, 95%CI 1.1-7.5), and having > 2 relatives with PC (OR 2.7, 95%CI 1.1-6.4) were independently associated with detection of pre-malignant and malignant lesions. The diagnostic yield for MRCP/EUS was 24% for cystic lesions. The overall rate of surgery was 2.6% with nil mortality. DISCUSSION The rate of malignancies found in this cohort was high (2.6%). According to the International Cancer of the Pancreas Screening Consortium the screening goal achievement was high (1%).
Collapse
Affiliation(s)
- Salvatore Paiella
- General and Pancreatic Surgery Department, Pancreas Institute, University of Verona, Verona, Italy
| | - Gabriele Capurso
- Digestive and Liver Disease Unit, S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Giulia Martina Cavestro
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Butturini
- Pancreatic Surgery Unit, Casa di Cura Pederzoli Hospital, Peschiera del Garda, Italy
| | | | - Roberto Salvia
- General and Pancreatic Surgery Department, Pancreas Institute, University of Verona, Verona, Italy
| | - Marianna Signoretti
- Digestive and Liver Disease Unit, S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Stefano Crippa
- Pancreatic Surgery Unit, IRCCS Ospedale San Raffaele Scientific Institute, Vita Salute San Raffaele University, Milano, Italy
| | - Silvia Carrara
- Gastrointestinal Endoscopy, Istituto Clinico Humanitas, Milano, Italy
| | - Isabella Frigerio
- Pancreatic Surgery Unit, Casa di Cura Pederzoli Hospital, Peschiera del Garda, Italy
| | - Claudio Bassi
- General and Pancreatic Surgery Department, Pancreas Institute, University of Verona, Verona, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, IRCCS Ospedale San Raffaele Scientific Institute, Vita Salute San Raffaele University, Milano, Italy
| | - Elsa Iannicelli
- Radiology Unit, S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Alessandro Giardino
- Pancreatic Surgery Unit, Casa di Cura Pederzoli Hospital, Peschiera del Garda, Italy
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Ospedale San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Laghi
- Radiology Unit, S. Andrea Hospital, University Sapienza, Rome, Italy
| | - Luigi Laghi
- Hereditary Cancer Genetics Clinic, Humanitas Clinical and Research Center, Milano, Italy
| | - Luca Frulloni
- Gastroenterology B Unit, Pancreas Institute, University of Verona, Verona, Italy
| | - Alessandro Zerbi
- Pancreatic Surgery Unit, Humanitas Clinical and Research Center, Milano, Italy
| |
Collapse
|
16
|
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
|
17
|
Diaz KE, Lucas AL. Familial Pancreatic Ductal Adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:36-43. [PMID: 30558720 PMCID: PMC7073774 DOI: 10.1016/j.ajpath.2018.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/21/2018] [Accepted: 06/11/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), although a rare disease, has a poor prognosis. With 5-year overall survival of 8%, there is a critical need to detect PDAC early or at a premalignant stage. Current screening methods are largely imaging based, but a more focused screening approach based on modifiable and nonmodifiable risk factors may improve the efficacy and likely outcomes of screening. In addition, the pathologic mechanisms that lead to the development of PDAC are discussed in an effort to further understand the targets of pancreatic cancer screening. The focus of this article will be inherited pancreatic cancer syndromes and familial pancreatic cancer, which together compose up to 10% of PDAC. Understanding the methods and targets of PDAC screening in high-risk individuals may translate to improved morbidity and mortality.
Collapse
Affiliation(s)
- Kelly E Diaz
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aimee L Lucas
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
18
|
Chhoda A, Lu L, Clerkin BM, Risch H, Farrell JJ. Current Approaches to Pancreatic Cancer Screening. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:22-35. [PMID: 30558719 DOI: 10.1016/j.ajpath.2018.09.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/29/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of only 8% and is estimated to be the second leading cause of cancer-related deaths by 2021. Prior convention held that screening for PDAC would not be beneficial; however, a deeper understanding of the carcinogenesis pathway supports a potential window of opportunity among the target population. Screening for PDAC is not a standard practice among the general population because of its low incidence. However, screening may be beneficial for individuals with familial history, chronic diseases with genetic predispositions, or inherited cancer syndromes, such as hereditary breast ovarian cancer syndrome, hereditary pancreatitis, Peutz-Jeghers syndrome, familial atypical multiple mole melanoma, Lynch syndrome (hereditary nonpolyposis colorectal cancer), ataxia telangiectasia, and Li-Fraumeni syndrome, all of which have been associated with an increased risk of developing PDAC. The screening strategies among these high-risk individuals are targeted to identify precursor lesions and PDAC at an early resectable stage. This review describes the risk factors for pancreatic cancer, especially the genetic risk factors in high-risk individuals and current screening strategies available for PDAC.
Collapse
Affiliation(s)
- Ankit Chhoda
- Yale Waterbury Internal Medicine Program, Yale School of Medicine, New Haven, Connecticut
| | - Lingeng Lu
- Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Barbara M Clerkin
- Pancreatic Disease Program, Yale School of Medicine, New Haven, Connecticut
| | - Harvey Risch
- Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - James J Farrell
- Yale Center for Pancreatic Diseases, Yale School of Medicine, New Haven, Connecticut; Yale Center for Pancreatic Diseases, Department of Digestive Diseases, Yale School of Public Health, New Haven, Connecticut.
| |
Collapse
|
19
|
Molina-Montes E, Malats N. Response to: Variation of the age at onset of pancreatic cancer according to tobacco smoking and family history. Int J Epidemiol 2018; 47:1358-1359. [PMID: 29800296 DOI: 10.1093/ije/dyy085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Esther Molina-Montes
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, CIBERONC, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO), Madrid, CIBERONC, Spain
| | | |
Collapse
|
20
|
Welinsky S, Lucas AL. Familial Pancreatic Cancer and the Future of Directed Screening. Gut Liver 2018; 11:761-770. [PMID: 28609837 PMCID: PMC5669591 DOI: 10.5009/gnl16414] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer (PC) is the third most common cause of cancer-related death in the United States and the 12th most common worldwide. Mortality is high, largely due to late stage of presentation and suboptimal treatment regimens. Approximately 10% of PC cases have a familial basis. The major genetic defect has yet to be identified but may be inherited by an autosomal dominant pattern with reduced penetrance. Several known hereditary syndromes or genes are associated with an increased risk of developing PC and account for approximately 2% of PCs. These syndromes include the hereditary breast-ovarian cancer syndrome, Peutz-Jeghers syndrome, familial atypical multiple mole melanoma, Lynch syndrome, familial polyposis, ataxia-telangiectasia, and hereditary pancreatitis. Appropriate screening using methods such as biomarkers or imaging, with endoscopic ultrasound and magnetic resonance imaging, may assist in the early detection of neoplastic lesions in the high-risk population. If these lesions are detected and treated before the development of invasive carcinoma, PC disease morbidity and mortality may be improved. This review will focus on familial PC and other hereditary syndromes implicated in the increased risk of PC; it will also highlight current screening methods and the future of new screening modalities.
Collapse
Affiliation(s)
- Sara Welinsky
- Samuel F. Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aimee L Lucas
- Samuel F. Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Endoscopic Ultrasound-Based Pancreatic Cancer Screening of High-Risk Individuals: A Prospective Observational Trial. Pancreas 2018; 47:586-591. [PMID: 29683970 DOI: 10.1097/mpa.0000000000001038] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Pancreatic cancer (PC), a common cause of cancer death, is rarely diagnosed at an early stage. Early detection of PC may improve outcomes in affected patients. This study evaluated the utility of screening of high-risk individuals (HRIs) using an endoscopic ultrasound (EUS)-only approach to detect early malignant changes. METHODS A prospective PC screening program for HRIs was opened in 2007. Fifty-eight patients have enrolled to date. Patients with normal EUS examinations underwent repeat EUS annually for 5 years. Patients with abnormal EUS underwent fine-needle aspiration (FNA) if a mass/cyst 1 cm or longer was found. Those with cysts/mass shorter than 1 cm or benign FNA underwent repeat EUS in 3 months. If unchanged, patients were followed with magnetic resonance imaging. RESULTS Thirty-nine patients (67%) had initial normal EUS examinations, and 16 patients completed the 5-year trial. Five patients who initially had a normal EUS developed cysts on subsequent examinations. Of the 24 subjects (41%) with abnormal findings, 3 underwent FNA: 2 consistent with intraductal papillary mucinous neoplasm, 1 with benign cytology. The 21 remaining patients had 1 subcentimeter cyst or more followed by magnetic resonance imaging. No PCs have been detected. CONCLUSIONS Precancerous cysts are frequently detected with EUS in HRI. Whether screening impacts survival in HRIs remains unclear and requires further evaluation in larger multicenter trials.
Collapse
|
22
|
Tezuka K, Ishiyama T, Takeshita A, Matsumoto H, Jingu A, Kikuchi J, Yamaya H, Ohe R, Ishizawa T. Poorly differentiated ductal adenocarcinoma of the pancreas with rapid progression in a young man. Clin J Gastroenterol 2018; 11:417-423. [PMID: 29663140 DOI: 10.1007/s12328-018-0859-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 04/06/2018] [Indexed: 10/17/2022]
Abstract
Pancreatic cancer in young adults is very rare. We report a case of young-onset poorly differentiated pancreatic ductal adenocarcinoma with rapid progression and poor prognosis in a 31-year-old Japanese man with no obvious family history of malignancy. Preoperative examinations revealed a mass lesion in the body of the pancreas, accompanied by a slightly dilated main pancreatic duct distal to the mass lesion. Pancreatic cancer with acute pancreatitis was suspected because of an elevation of serum pancreatic enzyme and tumor marker, along with imaging findings. Distal pancreatectomy with resection of the common hepatic artery and splenectomy along with lymph node dissection was performed. Microscopically, the tumor was mainly composed of poorly differentiated ductal adenocarcinoma. The postoperative course was uneventful, but the patient had multiple liver metastases 2 months postoperatively, in spite of adjuvant chemotherapy, and died 8 months postoperatively. This case may represent a rare instance of young-onset poorly differentiated ductal adenocarcinoma with rapid progression and may indicate potential risk factors of pancreatic cancer in young adults.
Collapse
Affiliation(s)
- Koji Tezuka
- Department of Surgery, Yamagata Prefectural Shinjo Hospital, 12-55 Wakabacho, Shinjo, 996-0025, Japan
| | - Tomoharu Ishiyama
- Department of Surgery, Yamagata Prefectural Shinjo Hospital, 12-55 Wakabacho, Shinjo, 996-0025, Japan.
| | - Akiko Takeshita
- Department of Surgery, Okitama Public General Hospital, 2000 Nishi-Otsuka, Higashi-Okitama-gun, Kawanishi, Yamagata, 992-0601, Japan
| | - Hidekazu Matsumoto
- Department of Surgery, Yamagata Prefectural Shinjo Hospital, 12-55 Wakabacho, Shinjo, 996-0025, Japan
| | - Akira Jingu
- Department of Surgery, Yamagata Prefectural Shinjo Hospital, 12-55 Wakabacho, Shinjo, 996-0025, Japan
| | - Jiro Kikuchi
- Department of Surgery, Yamagata Prefectural Shinjo Hospital, 12-55 Wakabacho, Shinjo, 996-0025, Japan
| | - Hideyuki Yamaya
- Department of Surgery, Yamagata Prefectural Shinjo Hospital, 12-55 Wakabacho, Shinjo, 996-0025, Japan
| | - Rintaro Ohe
- Department of Pathological Diagnostics, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Tetsuya Ishizawa
- Department of Gastroenterology, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| |
Collapse
|
23
|
Cremin C, Howard S, Le L, Karsan A, Schaeffer DF, Renouf D, Schrader KA. CDKN2A founder mutation in pancreatic ductal adenocarcinoma patients without cutaneous features of Familial Atypical Multiple Mole Melanoma (FAMMM) syndrome. Hered Cancer Clin Pract 2018. [PMID: 29541281 PMCID: PMC5842519 DOI: 10.1186/s13053-018-0088-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Approximately 5% to 10% of pancreatic ductal adenocarcinoma (PDAC) has a hereditary basis. In most of these defined hereditary cancer syndromes, PDAC is not the predominant cancer type. Traditional criteria for publicly funded genetic testing typically require the presence of a set combination of the predominant syndrome-associated cancer types in the family history. We report the identification of a CDKN2A pathogenic variant in a PDAC-prone family without the cutaneous features of multiple moles or melanoma that are characteristic of the Familial Atypical Multiple Mole Melanoma (FAMMM) Syndrome identified in a universal testing algorithm for inherited mutations in pancreatic cancer patients. Case presentation We present the case of two brothers of English ancestry diagnosed with PDAC within the same 12 month period, at the respective ages of 63 and 64 years of age. Neither brother reported a personal history of multiple moles or melanoma. Family history was positive for two second-degree relatives diagnosed with PDAC but was negative for other cancers or multiple moles in first- and second-degree relatives. Due to the absence of melanoma, this family did not meet provincial criteria for publicly funded genetic testing. Clinical genetic testing offered through a research grant identified a pathogenic variant in the CDKN2A gene c.377 T > A (p.Val126Asp). This variant is a North American founder mutation believed to pre-date colonization. Conclusions This case reminds clinicians to consider the possibility of a germline CDKN2A mutation in families with a high prevalence of PDAC, even in the absence of moles or melanoma. This case supports recent guidelines published by the American College of Medical Genetics and Genomics (ACMG) that genetics referrals are indicated in families with three or more cases of PDAC regardless of other cancer types in the family. A multi-gene panel approach is of particular benefit in diagnosing inherited cancer susceptibility in PDAC-only families.
Collapse
Affiliation(s)
- Carol Cremin
- 1Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia Canada.,Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia Canada.,Pancreas Centre BC, Vancouver, British Columbia Canada
| | - Sarah Howard
- Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia Canada.,Pancreas Centre BC, Vancouver, British Columbia Canada
| | - Lyly Le
- Department of Medical Oncology, BC Cancer - Surrey, Surrey, British Columbia Canada
| | - Aly Karsan
- 4Centre for Clinical Genomics, Genome Sciences Centre, BC Cancer Research Centre, Vancouver, British Columbia Canada.,5Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia Canada
| | - David F Schaeffer
- Pancreas Centre BC, Vancouver, British Columbia Canada.,7Division of Anatomical Pathology, Vancouver General Hospital, Vancouver, British Columbia Canada
| | - Daniel Renouf
- Pancreas Centre BC, Vancouver, British Columbia Canada.,Division of Medical Oncology, BC Cancer, Vancouver, British Columbia Canada
| | - Kasmintan A Schrader
- 1Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia Canada.,Hereditary Cancer Program, BC Cancer, Vancouver, British Columbia Canada.,Pancreas Centre BC, Vancouver, British Columbia Canada.,9Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, British Columbia Canada
| |
Collapse
|
24
|
Puri A, Chang JC, Kundranda M. Screening for Pancreatic Cancer: Current Status and Future Directions. EUROPEAN MEDICAL JOURNAL 2017. [DOI: 10.33590/emj/10313242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is a lethal disease for a multitude of reasons, including difficulty of early detection, early metastatic spread, and absence of more effective therapies. Even with the advent of newer systemic therapies, the 1-year survival for metastatic disease ranges from 17–23% and 5-year survival is <5%. This necessitates an urgent need for the development of more effective modalities for early detection, particularly due to the long latent period between the genomic cellular changes and the development of metastatic disease. Currently available biochemical and molecular markers have significant potential; however, they require further clinical validation. Endoscopic ultrasound is one of the most sensitive modalities used to both screen and sample lesions, but is limited to use in high-risk patients due to its invasive nature and associated risks. Although clinically meaningful progress has been made in screening the high-risk cohorts in terms of detection of pancreatic ductal adenocarcinoma, intraductal papillary mucinous neoplasms, and mucinous cystic neoplasms, leading to early diagnosis and treatment, nonselective population-based screening is not yet available for widespread use. Currently there is no consensus on the most appropriate screening protocol for early pancreatic cancer detection. In this review, we focus on understanding the potential role of molecular and radiogenomic markers in the early detection of pancreatic cancer.
Collapse
Affiliation(s)
- Akshjot Puri
- College of Medicine, University of Arizona, Phoenix, Arizona, USA
| | - John C. Chang
- Department of Radiology, Banner MD Anderson Cancer Center, Gilbert, Arizona, USA
| | - Madappa Kundranda
- Division of Oncology, Banner MD Anderson Cancer Center, Gilbert, Arizona, USA
| |
Collapse
|
25
|
Moutinho-Ribeiro P, Coelho R, Giovannini M, Macedo G. Pancreatic cancer screening: Still a delusion? Pancreatology 2017; 17:754-765. [PMID: 28739291 DOI: 10.1016/j.pan.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/17/2017] [Accepted: 07/05/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic adenocarcinoma represents the fourth most common cause of cancer mortality and death due to pancreatic cancer (PC) have increased since 2003. Its incidence has also raised about 30% in the past decade and it is expected to become the second cause of cancer mortality by 2020 in the USA. Most PC present with metastatic disease and improvements in treatment outcomes for this group have been disappointing. These observations support the idea that screening to identify patients at an earlier stage might be an important strategy in improving overall PC outcomes. Many protocols have been tested, nevertheless, by now there is no effective screening program. Given the overall low incidence of disease and the current lack of accurate, inexpensive and noninvasive screening tests, the consensus is that widespread population-based screening for PC in the general population or in patients with only one affected first-degree relative is neither practicable nor indicated in most countries. However, a different scenario is screening patients with higher risk for PC, most of them with hereditary conditions predisposing the development of this neoplasia. In fact, some guidelines are now available helping to select these individuals at risk and to screen them, in order to achieve early detection of PC.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal.
| | - Rosa Coelho
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
| | - Marc Giovannini
- Endoscopic Unit, Paoli-Calmettes Institute, Marseilles, France
| | - Guilherme Macedo
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|
26
|
|
27
|
Abstract
Pancreatic cancer (PC) is a highly fatal disease that can only be cured by complete surgical resection. However, most patients with PC have unresectable disease at the time of diagnosis, highlighting the need to detect PC and its precursor lesions earlier in asymptomatic patients. Screening is not cost-effective for population-based screening of PC. Individuals with genetic risk factors for PC based on family history or known PC-associated genetic syndromes, however, can be a potential target for PC screening programs. This article provides an overview of the epidemiology and genetic background of familial PC and discusses diagnostic and management approaches.
Collapse
Affiliation(s)
- Saowanee Ngamruengphong
- Division of Gastroenterology and Hepatology, Johns Hopkins Hospital, Johns Hopkins Medical Institutions, Blalock 407, Baltimore, MD 21287, USA
| | - Marcia Irene Canto
- Division of Gastroenterology and Hepatology, Johns Hopkins Hospital, Johns Hopkins Medical Institutions, Blalock 407, Baltimore, MD 21287, USA.
| |
Collapse
|
28
|
Xue J, Zhao Q, Sharma V, Nguyen LP, Lee YN, Pham KL, Edderkaoui M, Pandol SJ, Park W, Habtezion A. Aryl Hydrocarbon Receptor Ligands in Cigarette Smoke Induce Production of Interleukin-22 to Promote Pancreatic Fibrosis in Models of Chronic Pancreatitis. Gastroenterology 2016; 151:1206-1217. [PMID: 27769811 PMCID: PMC5499510 DOI: 10.1053/j.gastro.2016.09.064] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 08/18/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Cigarette smoke has been identified as an independent risk factor for chronic pancreatitis (CP). Little is known about the mechanisms by which smoking promotes development of CP. We assessed the effects of aryl hydrocarbon receptor (AhR) ligands found in cigarette smoke on immune cell activation in humans and pancreatic fibrosis in animal models of CP. METHODS We obtained serum samples from patients with CP treated at Stanford University hospital and healthy individuals (controls) and isolated CD4+ T cells. Levels of interleukin-22 (IL22) were measured by enzyme-linked immunosorbent assay and smoking histories were collected. T cells from healthy nonsmokers and smokers were stimulated and incubated with AhR agonists (2,3,7,8-tetrachlorodibenzo-p-dioxin or benzo[a]pyrene) or antagonists and analyzed by flow cytometry. Mice were given intraperitoneal injections of caerulein or saline, with or without lipopolysaccharide, to induce CP. Some mice were given intraperitoneal injections of AhR agonists at the start of caerulein injection, with or without an antibody against IL22 (anti-IL22) starting 2 weeks after the first caerulein injection, or recombinant mouse IL22 or vehicle (control) intraperitoneally 4 weeks after the first caerulein injection. Mice were exposed to normal air or cigarette smoke for 6 h/d for 7 weeks and expression of AhR gene targets was measured. Pancreata were collected from all mice and analyzed by histology and quantitative reverse transcription polymerase chain reaction. Pancreatic stellate cells and T cells were isolated and studied using immunoblot, immunofluorescence, flow cytometry, and enzyme-linked immunosorbent analyses. RESULTS Mice given AhR agonists developed more severe pancreatic fibrosis (based on decreased pancreas size, histology, and increased expression of fibrosis-associated genes) than mice not given agonists after caerulein injection. In mice given saline instead of caerulein, AhR ligands did not induce fibrosis. Pancreatic T cells from mice given AhR agonists and caerulein were activated and expressed IL22, but not IL17 or interferon gamma. Human T cells exposed to AhR agonists up-regulated expression of IL22. In mice given anti-IL22, pancreatic fibrosis did not progress, whereas mice given recombinant IL22 had a smaller pancreas and increased fibrosis. Pancreatic stellate cells isolated from mouse and human pancreata expressed the IL22 receptor IL22RA1. Incubation of the pancreatic stellate cells with IL22 induced their expression of the extracellular matrix genes fibronectin 1 and collagen type I α1 chain, but not α2 smooth muscle actin or transforming growth factor-β. Serum samples from smokers had significantly higher levels of IL22 than those from nonsmokers. CONCLUSIONS AhR ligands found in cigarette smoke increase the severity of pancreatic fibrosis in mouse models of pancreatitis via up-regulation of IL22. This pathway might be targeted for treatment of CP and serve as a biomarker of disease.
Collapse
MESH Headings
- Actins/genetics
- Animals
- Antibodies/pharmacology
- Benzo(a)pyrene/pharmacology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Ceruletide
- Collagen Type I/genetics
- Collagen Type I, alpha 1 Chain
- Disease Models, Animal
- Fibronectins/genetics
- Fibrosis
- Gene Expression/drug effects
- Humans
- Interferon-gamma/metabolism
- Interleukin-17/metabolism
- Interleukins/genetics
- Interleukins/immunology
- Interleukins/metabolism
- Interleukins/pharmacology
- Ligands
- Lymphocyte Activation
- Mice
- Pancreas/pathology
- Pancreatic Stellate Cells/chemistry
- Pancreatitis, Chronic/chemically induced
- Pancreatitis, Chronic/immunology
- Pancreatitis, Chronic/metabolism
- Pancreatitis, Chronic/pathology
- Polychlorinated Dibenzodioxins/pharmacology
- Receptors, Aryl Hydrocarbon/agonists
- Receptors, Aryl Hydrocarbon/antagonists & inhibitors
- Receptors, Interleukin/analysis
- Smoke
- Smoking/immunology
- Tobacco Products
- Transforming Growth Factor beta/genetics
- Interleukin-22
Collapse
Affiliation(s)
- Jing Xue
- State Key Laboratory of Oncogenes and Related Genes, Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California.
| | - Qinglan Zhao
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Vishal Sharma
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Linh P Nguyen
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Yvonne N Lee
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Kim L Pham
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Mouad Edderkaoui
- Cedars-Sinai Medical Center and Department of Veterans Affairs, Los Angeles, California
| | - Stephen J Pandol
- Cedars-Sinai Medical Center and Department of Veterans Affairs, Los Angeles, California
| | - Walter Park
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
29
|
Barone E, Corrado A, Gemignani F, Landi S. Environmental risk factors for pancreatic cancer: an update. Arch Toxicol 2016; 90:2617-2642. [PMID: 27538405 DOI: 10.1007/s00204-016-1821-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PC) is one of the most aggressive diseases. Only 10 % of all PC cases are thought to be due to genetic factors. Here, we analyzed the most recently published case-control association studies, meta-analyses, and cohort studies with the aim to summarize the main environmental factors that could have a role in PC. Among the most dangerous agents involved in the initiation phase, there are the inhalation of cigarette smoke, and the exposure to mutagenic nitrosamines, organ-chlorinated compounds, heavy metals, and ionizing radiations. Moreover, pancreatitis, high doses of alcohol drinking, the body microbial infections, obesity, diabetes, gallstones and/or cholecystectomy, and the accumulation of asbestos fibers seem to play a crucial role in the progression of the disease. However, some of these agents act both as initiators and promoters in pancreatic acinar cells. Protective agents include dietary flavonoids, marine omega-3, vitamin D, fruit, vegetables, and the habit of regular physical activity. The identification of the factors involved in PC initiation and progression could be of help in establishing novel therapeutic approaches by targeting the molecular signaling pathways responsive to these stimuli. Moreover, the identification of these factors could facilitate the development of strategies for an early diagnosis or measures of risk reduction for high-risk people.
Collapse
Affiliation(s)
- Elisa Barone
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy
| | - Alda Corrado
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy
| | - Federica Gemignani
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy
| | - Stefano Landi
- Genetic Unit, Department of Biology, University of Pisa, Via Derna, 1, 56121, Pisa, Italy.
| |
Collapse
|
30
|
Camara SN, Yin T, Yang M, Li X, Gong Q, Zhou J, Zhao G, Yang ZY, Aroun T, Kuete M, Ramdany S, Camara AK, Diallo AT, Feng Z, Ning X, Xiong JX, Tao J, Qin Q, Zhou W, Cui J, Huang M, Guo Y, Gou SM, Wang B, Liu T, Olivier OET, Conde T, Cisse M, Magassouba AS, Ballah S, Keita NLM, Souare IS, Toure A, Traore S, Balde AK, Keita N, Camara ND, Emmanuel D, Wu HS, Wang CY. High risk factors of pancreatic carcinoma. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2016; 36:295-304. [PMID: 27376795 DOI: 10.1007/s11596-016-1583-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/15/2016] [Indexed: 12/20/2022]
Abstract
Over the past decades, cancer has become one of the toughest challenges for health professionals. The epidemiologists are increasingly directing their research efforts on various malignant tumor worldwide. Of note, incidence of cancers is on the rise more quickly in developed countries. Indeed, great endeavors have to be made in the control of the life-threatening disease. As we know it, pancreatic cancer (PC) is a malignant disease with the worst prognosis. While little is known about the etiology of the PC and measures to prevent the condition, so far, a number of risk factors have been identified. Genetic factors, pre-malignant lesions, predisposing diseases and exogenous factors have been found to be linked to PC. Genetic susceptibility was observed in 10% of PC cases, including inherited PC syndromes and familial PC. However, in the remaining 90%, their PC might be caused by genetic factors in combination with environmental factors. Nonetheless, the exact mechanism of the two kinds of factors, endogenous and exogenous, working together to cause PC remains poorly understood. The fact that most pancreatic neoplasms are diagnosed at an incurable stage of the disease highlights the need to identify risk factors and to understand their contribution to carcinogenesis. This article reviews the high risk factors contributing to the development of PC, to provide information for clinicians and epidemiologists.
Collapse
Affiliation(s)
- Soriba Naby Camara
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Yin
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ming Yang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Li
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiong Gong
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gang Zhao
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhi-Yong Yang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tajoo Aroun
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Martin Kuete
- Department of Planning Family and Reproductive Institute, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sonam Ramdany
- Department of General Medicine, Sir Seewoosagur Ramgoolam National Hospital of Pamplemousses, Mauritius, 21017, Mauritius
| | | | - Aissatou Taran Diallo
- Department of General Surgery, National Hospital of Ignace Deen, Conakry, 1147, Guinea
| | - Zhen Feng
- Department of Gastroenterology and Hepatology, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xin Ning
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiong-Xin Xiong
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Tao
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qi Qin
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Zhou
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jing Cui
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Huang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yao Guo
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shan-Miao Gou
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Wang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Liu
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ohoya Etsaka Terence Olivier
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tenin Conde
- Department of Thoracic Surgery, National Hospital of Donka, Conakry, Guinea
| | - Mohamed Cisse
- Department of Dermatology, National Hospital of Donka, Conakry, Guinea
| | | | - Sneha Ballah
- Department of Internal Medicine, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Naby Laye Moussa Keita
- Department of Biochemistry, University Gamal Abdel Nasser of Conakry, Conakry, 1147, Guinea
| | - Ibrahima Sory Souare
- Department of Neurosurgery, Friendship Hospital Sino-Guinea of Kipe, Conakry, Guinea
| | - Aboubacar Toure
- Department of General Surgery, National Hospital of Ignace Deen, Conakry, 1147, Guinea
| | - Sadamoudou Traore
- Department of Medical Imaging, Good Shepherd Medical Center, The University of Texas, Longview, 75601, USA
| | | | - Namory Keita
- Department of Gynecology and Obstetrics, National Hospital of Donka, Conakry, Guinea
| | - Naby Daouda Camara
- Department of General Surgery, National Hospital of Ignace Deen, Conakry, 1147, Guinea
| | - Dusabe Emmanuel
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - He-Shui Wu
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun-You Wang
- Department of General Surgery, Pancreatic Disease Institute, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
31
|
|
32
|
Joergensen MT, Gerdes AM, Sorensen J, Schaffalitzky de Muckadell O, Mortensen MB. Is screening for pancreatic cancer in high-risk groups cost-effective? - Experience from a Danish national screening program. Pancreatology 2016; 16:584-92. [PMID: 27090585 DOI: 10.1016/j.pan.2016.03.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/17/2016] [Accepted: 03/19/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Pancreatic cancer (PC) is the fourth leading cause of cancer death worldwide, symptoms are few and diffuse, and when the diagnosis has been made only 10-15% would benefit from resection. Surgery is the only potentially curable treatment for pancreatic cancer, and the prognosis seems to improve with early detection. A hereditary component has been identified in 1-10% of the PC cases. To comply with this, screening for PC in high-risk groups with a genetic disposition for PC has been recommended in research settings. DESIGN Between January 2006 and February 2014 31 patients with Hereditary pancreatitis or with a disposition of HP and 40 first-degree relatives of patients with Familial Pancreatic Cancer (FPC) were screened for development of Pancreatic Ductal Adenocarcinoma (PDAC) with yearly endoscopic ultrasound. The cost-effectiveness of screening in comparison with no-screening was assessed by the incremental cost-utility ratio (ICER). RESULTS By screening the FPC group we identified 2 patients with PDAC who were treated by total pancreatectomy. One patient is still alive, while the other died after 7 months due to cardiac surgery complications. Stratified analysis of patients with HP and FPC provided ICERs of 47,156 US$ vs. 35,493 US$ per life-year and 58,647 US$ vs. 47,867 US$ per QALY. Including only PDAC related death changed the ICER to 31,722 US$ per life-year and 42,128 US$ per QALY. The ICER for patients with FPC was estimated at 28,834 US$ per life-year and 38,785 US$ per QALY. CONCLUSIONS With a threshold value of 50,000 US$ per QALY this screening program appears to constitute a cost-effective intervention although screening of HP patients appears to be less cost-effective than FPC patients.
Collapse
Affiliation(s)
- Maiken Thyregod Joergensen
- Vejle Hospital, Southern Denmark, Odense, Denmark; Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.
| | | | - Jan Sorensen
- Centre for Health Economic Research (COHERE), Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
33
|
Abstract
Pancreatic adenocarcinoma is a leading cause of cancer death. Few patients are candidates for curative resection due to the late stage at diagnosis. While most pancreatic adenocarcinomas are sporadic, approximately 10% have an underlying hereditary basis. Known genetic syndromes account for only 20% of the familial clustering of pancreatic cancer cases. The majority are due to non-syndromic aggregation of pancreatic cancer cases or familial pancreatic cancer. Screening aims to identify high-risk lesions amenable to surgical resection. However, the optimal interval for screening and the management of pancreatic cancer precursor lesions detected on imaging are controversial.
Collapse
Affiliation(s)
- Shilpa Grover
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Kunal Jajoo
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
34
|
Risk Factors for Early-Onset and Very-Early-Onset Pancreatic Adenocarcinoma: A Pancreatic Cancer Case-Control Consortium (PanC4) Analysis. Pancreas 2016; 45:311-6. [PMID: 26646264 PMCID: PMC4710562 DOI: 10.1097/mpa.0000000000000392] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES While pancreatic cancer (PC) most often affects older adults, to date, there has been no comprehensive assessment of risk factors among PC patients younger than 60 years. METHODS We defined early-onset PC (EOPC) and very-early-onset PC (VEOPC) as diagnosis of PC in patients younger than 60 and 45 years, respectively. We pooled data from 8 case-control studies, including 1954 patients with EOPC and 3278 age- and sex-matched control subjects. Logistic regression analysis was performed to identify associations with EOPC and VEOPC. RESULTS Family history of PC, diabetes mellitus, smoking, obesity, and pancreatitis were associated with EOPC. Alcohol use equal to or greater than 26 g daily also was associated with increased risk of EOPC (odds ratio, 1.49; 95% confidence interval, 1.21-1.84), and there appeared to be a dose- and age-dependent effect of alcohol on risk. The point estimate for risk of VEOPC was an odds ratio of 2.18 (95% confidence interval, 1.17-4.09). CONCLUSIONS The established risk factors for PC, including smoking, diabetes, family history of PC, and obesity, also apply to EOPC. Alcohol intake appeared to have an age-dependent effect; the strongest association was with VEOPC.
Collapse
|
35
|
Bhutani MS, Koduru P, Joshi V, Saxena P, Suzuki R, Irisawa A, Yamao K. The role of endoscopic ultrasound in pancreatic cancer screening. Endosc Ultrasound 2016; 5:8-16. [PMID: 26879161 PMCID: PMC4770628 DOI: 10.4103/2303-9027.175876] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 10/13/2015] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer (PC) is a highly lethal cancer. Despite a significant advancement in cancer treatment, the mortality rate of PC is nearly identical to the incidence rates. Early detection of tumor or its precursor lesions with dysplasia may be the most effective approach to improve survival. Screening strategies should include identification of the population at high risk of developing PC, and an intense application of screening tools with adequate sensitivity to detect PC at an early curable stage. Endoscopic ultrasound (EUS) and magnetic resonance imaging (MRI) seem to be the most promising modalities for PC screening based on the data so far. EUS had an additional advantage over MRI by being able to obtain tissue sample during the same examination. Several questions remain unanswered at this time regarding the age to begin screening, frequency of screening, management of asymptomatic pancreatic lesions detected on screening, timing of resection, and extent of surgery and impact of screening on survival. Novel techniques such as needle-based confocal laser endomicroscopy (nCLE), along with biomarkers, may be helpful to identify pancreatic lesions with more aggressive malignant potential. Further studies will hopefully lead to the development of strategies combining EUS with other technological/biological advancements that will be cost-effective and have an impact on survival.
Collapse
Affiliation(s)
- Manoop S. Bhutani
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Pramoda Koduru
- Department of Gastroenterology, Hepatology and Nutrition, MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Virendra Joshi
- Department of Gastroenterology, Ochsner Clinic Foundation, Ochsner Cancer Institute, New Orleans, Louisiana, USA
| | - Payal Saxena
- Department of Gastroenterology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Rei Suzuki
- Department of Gastroenterology and Rheumatology, Fukushima Medical University School of Medicine, Aizuwakamatsu, Fukushima, Japan
| | - Atsushi Irisawa
- Department of Gastroenterology, Aizu Medical Center, Fukushima Medical University, Aizuwakamatsu, Fukushima, Japan
| | - Kenji Yamao
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| |
Collapse
|
36
|
Huang J, Löhr JM, Nilsson M, Segersvärd R, Matsson H, Verbeke C, Heuchel R, Kere J, Iafrate AJ, Zheng Z, Ye W. Variant Profiling of Candidate Genes in Pancreatic Ductal Adenocarcinoma. Clin Chem 2015; 61:1408-1416. [PMID: 26378065 DOI: 10.1373/clinchem.2015.238543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 08/28/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Variant profiling is crucial for developing personalized treatment and elucidating the etiology of this disease. METHODS Patients with PDAC undergoing surgery from 2007 to 2012 (n = 73) were followed from diagnosis until death or the end of the study. We applied an anchored multiplex PCR (AMP)-based next-generation sequencing (NGS) method to a panel of 65 selected genes and assessed analytical performance by sequencing a quantitative multiplex DNA reference standard. In clinical PDAC samples, detection of low-level KRAS (Kirsten rat sarcoma viral oncogene homolog) mutations was validated by allele-specific PCR and digital PCR. We compared overall survival of patients according to KRAS mutation status by log-rank test and applied logistic regression to evaluate the association between smoking and tumor variant types. RESULTS The AMP-based NGS method could detect variants with allele frequencies as low as 1% given sufficient sequencing depth (>1500×). Low-frequency KRAS G12 mutations (allele frequency 1%-5%) were all confirmed by allele-specific PCR and digital PCR. The most prevalent genetic alterations were in KRAS (78% of patients), TP53 (tumor protein p53) (25%), and SMAD4 (SMAD family member 4) (8%). Overall survival in T3-stage PDAC patients differed among KRAS mutation subtypes (P = 0.019). Transversion variants were more common in ever-smokers than in never-smokers (odds ratio 5.7; 95% CI 1.2-27.8). CONCLUSIONS The AMP-based NGS method is applicable for profiling tumor variants. Using this approach, we demonstrated that in PDAC patients, KRAS mutant subtype G12V is associated with poorer survival, and that transversion variants are more common among smokers.
Collapse
Affiliation(s)
- Jiaqi Huang
- Department of Medical Epidemiology and Biostatistics and
| | | | - Magnus Nilsson
- Center for Digestive Diseases, Division of Surgery, CLINTEC, and
| | - Ralf Segersvärd
- Center for Digestive Diseases, Division of Surgery, CLINTEC, and
| | - Hans Matsson
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Huddinge, Sweden
| | - Caroline Verbeke
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Clinical Pathology/Cytology, Karolinska University Hospital, Stockholm, Sweden
| | - Rainer Heuchel
- Center for Digestive Diseases, Division of Surgery, CLINTEC, and
| | - Juha Kere
- Department of Biosciences and Nutrition and Center for Innovative Medicine (CIMED), Karolinska Institutet, Huddinge, Sweden
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Zongli Zheng
- Department of Medical Epidemiology and Biostatistics and Department of Pathology, Massachusetts General Hospital, Boston, MA; Current address: Department of Biomedical Sciences, City University of Hong Kong, Hong Kong.
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics and
| |
Collapse
|
37
|
Abstract
Despite decades of scientific and clinical research, pancreatic ductal adenocarcinoma (PDAC) remains a lethal malignancy. The clinical and pathologic features of PDAC, specifically the known environmental and genetic risk factors, are reviewed here with special emphasis on the hereditary pancreatic cancer (HPC) syndromes. For these latter conditions, strategies are described for their identification, for primary and secondary prevention in unaffected carriers, and for disease management in affected carriers. Nascent steps have been made toward personalized medicine based on the rational use of screening, tumor subtyping, and targeted therapies; these have been guided by growing knowledge of HPC syndromes in PDAC.
Collapse
Affiliation(s)
- Ashton A Connor
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Gallinger
- Division of General Surgery, Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Mocci E, Guillen-Ponce C, Earl J, Marquez M, Solera J, Salazar-López MT, Calcedo-Arnáiz C, Vázquez-Sequeiros E, Montans J, Muñoz-Beltrán M, Vicente-Bártulos A, González-Gordaliza C, Sanjuanbenito A, Guerrero C, Mendía E, Lisa E, Lobo E, Martínez JC, Real FX, Malats N, Carrato A. PanGen-Fam: Spanish registry of hereditary pancreatic cancer. Eur J Cancer 2015. [PMID: 26212471 DOI: 10.1016/j.ejca.2015.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE To describe the organisation of the registry and the preliminary results in terms of characteristics of high-risk pancreatic ductal adenocarcinoma (PDAC) families recruited to date and findings of the screening programme. To compare early onset sporadic cases (⩽50 years), sporadic cases (>50 years) and cases with family history of cancer, for PDAC possible risk factors. METHODS/PATIENTS Families with hereditary cancer syndromes predisposing to PDAC were recruited from two main sources: Spanish hospitals participating in PanGenEU, a pan-European multicentre case-control study, and their genetic counseling unit. Individuals at high-risk of PDAC were enrolled into a screening programme, consisting of Endoscopic ultrasound, computerised tomography, magnetic resonance imaging. Genetic testing of candidate genes was offered according to each patient's risk. RESULTS Among 577 consecutive PDAC cases, recruited via PanGenEU, 36 (6%) had ⩾2 first-degree relative with PDAC: Familial pancreatic cancer (FPC). So far PanGen-Fam has recruited 42 high-risk PDAC families; 25 (60%) had FPC. Five index cases with cancer were positive for BRCA2 and one for BRCA1 germline mutations. In the second year of prospective PDAC screening, one neuroendocrine tumour and a high-grade dysplasia lesion suspicious of carcinoma were diagnosed among 41 high-risk individuals. Furthermore EUS detected chronic-pancreatitis-like parenchymal changes in 15 patients. CONCLUDING STATEMENT The identification and recruitment of PDAC high-risk families into the PanGen-Fam registry provides an opportunity to detect early onset cancer and precursor pancreatic cancer lesions at a potentially curative stage and to increase the knowledge of the natural history of the disease.
Collapse
Affiliation(s)
- E Mocci
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - C Guillen-Ponce
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - J Earl
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - M Marquez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - J Solera
- Molecular Oncogenetics Unit, Institute of Medical and Molecular Genetics, La Paz Hospital, Madrid, Spain
| | - M-T Salazar-López
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - C Calcedo-Arnáiz
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - E Vázquez-Sequeiros
- Digestive Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - J Montans
- Pathology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - M Muñoz-Beltrán
- Radiology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - A Vicente-Bártulos
- Radiology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | | | - A Sanjuanbenito
- Surgery Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - C Guerrero
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - E Mendía
- Surgery Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - E Lisa
- Surgery Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - E Lobo
- Surgery Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| | - J C Martínez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - F X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain; Department de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - N Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - A Carrato
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, Madrid, Spain
| |
Collapse
|
39
|
Urayama S. Pancreatic cancer early detection: Expanding higher-risk group with clinical and metabolomics parameters. World J Gastroenterol 2015; 21:1707-1717. [PMID: 25684935 PMCID: PMC4323446 DOI: 10.3748/wjg.v21.i6.1707] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 10/01/2014] [Accepted: 01/08/2015] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth and fifth leading cause of cancer death for each gender in developed countries. With lack of effective treatment and screening scheme available for the general population, the mortality rate is expected to increase over the next several decades in contrast to the other major malignancies such as lung, breast, prostate and colorectal cancers. Endoscopic ultrasound, with its highest level of detection capacity of smaller pancreatic lesions, is the commonly employed and preferred clinical imaging-based PDAC detection method. Various molecular biomarkers have been investigated for characterization of the disease, but none are shown to be useful or validated for clinical utilization for early detection. As seen from studies of a small subset of familial or genetically high-risk PDAC groups, the higher yield and utility of imaging-based screening methods are demonstrated for these groups. Multiple recent studies on the unique cancer metabolism including PDAC, demonstrate the potential for utility of the metabolites as the discriminant markers for this disease. In order to generate an early PDAC detection screening strategy available for a wider population, we propose to expand the population of higher risk PDAC group with combination clinical and metabolomics parameters.
Collapse
|
40
|
ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes. Am J Gastroenterol 2015; 110:223-62; quiz 263. [PMID: 25645574 PMCID: PMC4695986 DOI: 10.1038/ajg.2014.435] [Citation(s) in RCA: 1082] [Impact Index Per Article: 108.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023]
Abstract
This guideline presents recommendations for the management of patients with hereditary gastrointestinal cancer syndromes. The initial assessment is the collection of a family history of cancers and premalignant gastrointestinal conditions and should provide enough information to develop a preliminary determination of the risk of a familial predisposition to cancer. Age at diagnosis and lineage (maternal and/or paternal) should be documented for all diagnoses, especially in first- and second-degree relatives. When indicated, genetic testing for a germline mutation should be done on the most informative candidate(s) identified through the family history evaluation and/or tumor analysis to confirm a diagnosis and allow for predictive testing of at-risk relatives. Genetic testing should be conducted in the context of pre- and post-test genetic counseling to ensure the patient's informed decision making. Patients who meet clinical criteria for a syndrome as well as those with identified pathogenic germline mutations should receive appropriate surveillance measures in order to minimize their overall risk of developing syndrome-specific cancers. This guideline specifically discusses genetic testing and management of Lynch syndrome, familial adenomatous polyposis (FAP), attenuated familial adenomatous polyposis (AFAP), MUTYH-associated polyposis (MAP), Peutz-Jeghers syndrome, juvenile polyposis syndrome, Cowden syndrome, serrated (hyperplastic) polyposis syndrome, hereditary pancreatic cancer, and hereditary gastric cancer.
Collapse
|
41
|
Humphris JL, Johns AL, Simpson SH, Cowley MJ, Pajic M, Chang DK, Nagrial AM, Chin VT, Chantrill LA, Pinese M, Mead RS, Gill AJ, Samra JS, Kench JG, Musgrove EA, Tucker KM, Spigelman AD, Waddell N, Grimmond SM, Biankin AV. Clinical and pathologic features of familial pancreatic cancer. Cancer 2014; 120:3669-75. [PMID: 25313458 DOI: 10.1002/cncr.28863] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/20/2014] [Accepted: 05/07/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Inherited predisposition to pancreatic cancer contributes significantly to its incidence and presents an opportunity for the development of early detection strategies. The genetic basis of predisposition remains unexplained in a high proportion of patients with familial PC (FPC). METHODS Clinicopathologic features were assessed in a cohort of 766 patients who had been diagnosed with pancreatic ductal adenocarcinoma (PC). Patients were classified with FPC if they had ≥1 affected first-degree relatives; otherwise, they were classified with sporadic PC (SPC). RESULTS The prevalence of FPC in this cohort was 8.9%. In FPC families with an affected parent-child pair, 71% in the subsequent generation were 12.3 years younger at diagnosis. Patients with FPC had more first-degree relatives who had an extrapancreatic malignancy (EPM) (42.6% vs 21.2; P<.0001), particularly melanoma and endometrial cancer, but not a personal history of EPM. Patients with SPC were more likely to be active smokers, have higher cumulative tobacco exposure, and have fewer multifocal precursor lesions, but these were not associated with differences in survival. Long-standing diabetes mellitus (>2 years) was associated with poor survival in both groups. CONCLUSIONS FPC represents 9% of PC, and the risk of malignancy in kindred does not appear to be confined to the pancreas. Patients with FPC have more precursor lesions and include fewer active smokers, but other clinicopathologic factors and outcome are similar to those in patients with SPC. Furthermore, some FPC kindreds may exhibit anticipation. A better understanding of the clinical features of PC will facilitate efforts to uncover novel susceptibility genes and the development of early detection strategies.
Collapse
Affiliation(s)
- Jeremy L Humphris
- The Kinghorn Cancer Center, Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Independent and joint effect of type 2 diabetes and gastric and hepatobiliary diseases on risk of pancreatic cancer risk: 10-year follow-up of population-based cohort. Br J Cancer 2014; 111:2180-6. [PMID: 25275365 PMCID: PMC4260037 DOI: 10.1038/bjc.2014.525] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/29/2014] [Accepted: 09/07/2014] [Indexed: 12/13/2022] Open
Abstract
Background: Type 2 diabetes mellitus, gastric and hepatobiliary comorbidities, and cancer share common risk factors: for example, tobacco, obesity, physical inactivity, high calorie intake, and metabolic disorders. Prior studies find type 2 diabetes and gastric and hepatobiliary comorbidities heightening risk of pancreatic cancer. Yet joint association of type 2 diabetes mellitus and gastric and hepatobiliary comorbidities on pancreatic cancer risk has not been assessed. Methods: This study rates independent/joint effects of type 2 diabetes as well as gastric and hepatobiliary comorbidity on pancreatic cancer risk for a retrospective population-based cohort of 166 850 type 2 diabetics identified in 1997–1998 and followed for 10–11 years, comparing their cancer incidence with that of 166 850 non-diabetics matched for age, gender, and locale. Time-dependent Cox's proportional hazards model evaluted joint association of type 2 diabetes and chronic conditions on pancreatic cancer risk. Results: A total of 1178 subjects were newly diagnosed with pancreatic cancer during follow-up, with incidence rates of 0.49 per 1000 person-years in type 2 diabetes and 0.26 per 1000 person-years in the non-diabetics. We observed greater magnitude of hazard ratios (HRs) of pancreatic cancer for patients with type 2 diabetes along with acute alcoholic hepatitis, acute pancreatitis, cholecystitis, and gastric ulcer compared with patients without type 2 diabetes or counterpart comorbidity (HR: 1.36, 95% confidence interval (CI): 1.19–1.56; 1.74, 1.23–2.45; 9.18, 7.44–11.33; and 2.31, 1.98–2.70, respectively). Main effects of type 2 diabetes were all statistically with narrow 95% CI and remained similar across risk stratification with various comorbidities: range 1.59–1.80. Conclusions: Our study demonstrates that pre-existing type 2 diabetes, acute alcoholic hepatitis, acute pancreatitis, cholecystitis, and gastric ulcer independently or jointly predict subsequent pancreatic cancer risk. Clinicians must recognise burden of these gastric and hepatobiliary comorbidities and keep clinically vigilant for their diagnosis.
Collapse
|
43
|
Mizuno S, Nakai Y, Isayama H, Kawahata S, Saito T, Takagi K, Watanabe T, Uchino R, Hamada T, Miyabayashi K, Kogure H, Sasaki T, Yamamoto N, Sasahira N, Hirano K, Tsujino T, Ijichi H, Tateishi K, Tada M, Koike K. Smoking, family history of cancer, and diabetes mellitus are associated with the age of onset of pancreatic cancer in Japanese patients. Pancreas 2014; 43:1014-7. [PMID: 24979618 DOI: 10.1097/mpa.0000000000000158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The aim of this study was to examine the association of risk factors including diabetes mellitus (DM) with the age of onset in Japanese pancreatic cancer (PC) patients. METHODS We retrospectively reviewed 688 PC patients diagnosed at our institute. We analyzed the association between the age of onset of PC and the following variables: sex, smoking, alcohol, DM, and a family history of cancer especially PC. RESULTS The mean age of PC diagnosis was 67.6 years. The onset of PC occurred earlier in current smokers (63.6 years old, P < 0.001) compared with past smokers (69.5 years old) and never smokers (68.6 years old). Patients with long-standing DM (>2 years) were older (70.5 years, P < 0.001) when diagnosed with PC than patients with new-onset DM (within 2 years) (66.9 years old) and patients without DM (66.7 years old). In the multivariate analysis, current smokers and a family history of cancer other than PC were associated with earlier onset. Conversely, long-standing DM was associated with later onset. CONCLUSIONS In Japanese PC patients, current smokers and a family history of cancer other than PC were associated with a younger age of onset. Conversely, long-standing DM was associated with a later onset.
Collapse
Affiliation(s)
- Suguru Mizuno
- From the Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Li H, Li Y, Cui L, Wang B, Cui W, Li M, Cheng Y. Monitoring pancreatic carcinogenesis by the molecular imaging of cathepsin E in vivo using confocal laser endomicroscopy. PLoS One 2014; 9:e106566. [PMID: 25184278 PMCID: PMC4153675 DOI: 10.1371/journal.pone.0106566] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/31/2014] [Indexed: 12/24/2022] Open
Abstract
The monitoring of pancreatic ductal adenocarcinoma (PDAC) in high-risk populations is essential. Cathepsin E (CTSE) is specifically and highly expressed in PDAC and pancreatic intraepithelial neoplasias (PanINs), and its expression gradually increases along with disease progression. In this study, we first established an in situ 7,12-dimethyl-1,2-benzanthracene (DMBA)-induced rat model for PanINs and PDAC and then confirmed that tumorigenesis properties in this model were consistent with those of human PDAC in that CTSE expression gradually increased with tumor development using histology and immunohistochemistry. Then, using in vivo imaging of heterotopically implanted tumors generated from CTSE- overexpressing cells (PANC-1-CTSE) in nude mice and in vitro imaging of PanINs and PDAC in DMBA-induced rats, the specificity of the synthesized CTSE-activatable probe was verified. Quantitative determination identified that the fluorescence signal ratio of pancreatic tumor to normal pancreas gradually increased in association with progressive pathological grades, with the exception of no significant difference between PanIN-II and PanIN-III grades. Finally, we monitored pancreatic carcinogenesis in vivo using confocal laser endomicroscopy (CLE) in combination with the CTSE-activatable probe. A prospective double-blind control study was performed to evaluate the accuracy of this method in diagnosing PDAC and PanINs of all grades (>82.7%). This allowed us to establish effective diagnostic criteria for CLE in PDAC and PanINs to facilitate the monitoring of PDAC in high-risk populations.
Collapse
Affiliation(s)
- Hui Li
- Department of Radiology, the Sixth Affiliated People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongdong Li
- Department of Radiology, the Sixth Affiliated People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Cui
- College of Science, University of Shanghai for Science and Technology, Shanghai, China
| | - Biyuan Wang
- Department of Radiology, the Sixth Affiliated People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenli Cui
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Minghua Li
- Department of Radiology, the Sixth Affiliated People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yingsheng Cheng
- Department of Radiology, the Sixth Affiliated People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
45
|
Batabyal P, Vander Hoorn S, Christophi C, Nikfarjam M. Association of diabetes mellitus and pancreatic adenocarcinoma: a meta-analysis of 88 studies. Ann Surg Oncol 2014; 21:2453-62. [PMID: 24609291 DOI: 10.1245/s10434-014-3625-6] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at an advanced, incurable stage. Previous epidemiological data suggests that diabetes mellitus (DM) is a risk factor for PDAC, which may be important in early detection. However, the strength of this association needs to be determined, taking into account a number of recently published studies. METHODS A systematic review of the association between DM and PDAC was undertaken by searching electronic databases and journal references from 1973 to 2013. Summary estimates were obtained separately for case-control and cohort studies by means of a 'random effects' approach. Data pertaining to the DM was recorded and plotted at both an individual and study level, with the relative risks (RR) pooled separately to determine the relationship of DM duration and PDAC. RESULTS A total of 88 independent studies, including 50 cohort and 39 case-control studies were examined. The overall summary-combined RR was 1.97 (95 % CI 1.78-2.18) with marked heterogeneity that could not be clearly attributed to any subgroup analyses. The risk of PDAC was greatest early after the diagnosis of DM but remained elevated long after the diagnosis. The individual-level RR ranged from 6.69 at less than 1 year to 1.36 at 10 years. CONCLUSION The results demonstrate a strong association between PDAC and recently diagnosed DM, which may be attributed to a paraneoplastic effect. However, the presence of diabetes also remains a modest risk factor for the development of PDAC long-term. Selective screening of patients with new-onset DM for PDAC needs to be considered.
Collapse
Affiliation(s)
- Pikli Batabyal
- Department of Surgery, University of Melbourne, Austin Health, LTB 8, 145 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | | | | | | |
Collapse
|
46
|
Reznik R, Hendifar AE, Tuli R. Genetic determinants and potential therapeutic targets for pancreatic adenocarcinoma. Front Physiol 2014; 5:87. [PMID: 24624093 PMCID: PMC3939680 DOI: 10.3389/fphys.2014.00087] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 02/13/2014] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer deaths in both men and women in the United States, carrying a 5-year survival rate of approximately 5%, which is the poorest prognosis of any solid tumor type. Given the dismal prognosis associated with PDAC, a more thorough understanding of risk factors and genetic predisposition has important implications not only for cancer prevention, but also for screening techniques and the development of personalized therapies. While screening of the general population is not recommended or practicable with current diagnostic methods, studies are ongoing to evaluate its usefulness in people with at least 5- to 10-fold increased risk of PDAC. In order to help identify high-risk populations who would be most likely to benefit from early detection screening tests for pancreatic cancer, discovery of additional pancreatic cancer susceptibility genes is crucial. Thus, specific gene-based, gene-product, and marker-based testing for the early detection of pancreatic cancer are currently being developed, with the potential for these to be useful as potential therapeutic targets as well. The goal of this review is to provide an overview of the genetic basis for PDAC with a focus on germline and familial determinants. A discussion of potential therapeutic targets and future directions in screening and treatment is also provided.
Collapse
Affiliation(s)
- Robert Reznik
- Department of Radiation Oncology, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Andrew E Hendifar
- Department of Radiation Oncology, Cedars-Sinai Medical Center Los Angeles, CA, USA
| | - Richard Tuli
- Department of Radiation Oncology, Cedars-Sinai Medical Center Los Angeles, CA, USA
| |
Collapse
|
47
|
Abstract
INTRODUCTION Familial pancreatic cancer (FPC) is defined by families with at least two first-degree relatives with confirmed pancreatic ductal adenocarcinoma (PDAC) that do not fulfill the criteria of other inherited tumor syndromes with an increased risk for the development of PDAC, such as hereditary pancreatitis or hereditary breast and ovarian cancer. FPC is mostly autosomal dominant inherited and presents with a heterogeneous phenotype. Although the major gene defect has not yet been identified, some important germline mutations in the BRCA2-, PALB2-, and ATM-genes are causative in some FPC families. FPC SCREENING It is suggested by experts to include high-risk individuals in a screening program with a multidisciplinary approach under research protocol conditions. However, neither biomarkers nor reliable imaging modalities for the detection of high-grade precursor lesions are yet available. Most screening programs are currently based on endoscopic ultrasound and magnetic resonance imaging, and first data demonstrated that precursor lesions (pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm) of PDAC can be identified. Timing and extent of surgery are still a matter of debate. SCOPE OF THE REVIEW The present review focuses on the clinical phenotype of FPC, its histopathological characteristics, known underlying genetic changes, genetic counseling, and screening.
Collapse
Affiliation(s)
- Volker Fendrich
- National Case Collection of Familial Pancreatic Cancer of the Deutsche Krebshilfe (FaPaCa), Department of Surgery, Philipps-University Marburg, Baldingerstrasse, 35043, Marburg, Germany,
| | | | | |
Collapse
|
48
|
Bouchbika Z, Haddad H, Benchakroun N, Eddakaoui H, Kotbi S, Megrini A, Bourezgui H, Sahraoui S, Corbex M, Harif M, Benider A. Cancer incidence in Morocco: report from Casablanca registry 2005-2007. Pan Afr Med J 2013; 16:31. [PMID: 24570792 PMCID: PMC3932129 DOI: 10.11604/pamj.2013.16.31.2791] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 07/16/2013] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Few population-based cancer registries are in place in developing countries. In order to know the burden of cancer in Moroccan population, cancer registry initiative was put in place in the Casablanca district, the biggest city of Morocco. METHODS The data collected covers 3.6 millions inhabitant and included Casablanca city and the administrative region. RESULTS The data collected in the years 2005-07 show that the top 5 forms of cancers in women were breast (ASR: 36.4 per 100,000), cervical (15.0), thyroid (6.7), colon-rectum (5.8), and ovarian (5.3); the top 5 cancers in men were lung (25.9), prostate (13.5), bladder (8.7), colon-rectum (8.1) and non-Hodgkin lymphoma (7.2). Tumours of haematopoietic and lymphoid tissues represented 11% of all cancers (skin excluded); some presented unusual sex ratios. For breast, cervical, colorectal and thyroid cancer, respectively 57%, 42%, 28% and 60% of the cases were under 50 years of age. This was attributable to particularly low numbers of cases recorded among old people, and the young age of the general population; the observed age-specific incidences under age 50 were not higher than in western countries. Cancers at young ages were particularly common in women: 67% of the cases were under 50. Stage at diagnosis could be obtained for 82% of the breast cancer cases and was as follows: 28% local, 63% regional and 9% distant, in the absence of screening. CONCLUSION These first population-based data have provided an invaluable resource for the national cancer control plan of Morocco, and will be useful tool to its future evaluation.
Collapse
Affiliation(s)
- Zineb Bouchbika
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| | - Houssam Haddad
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| | - Nadia Benchakroun
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| | - Houda Eddakaoui
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| | - Souad Kotbi
- Registry team, Regional Directorate of Health of Casablanca, Morocco
| | - Anis Megrini
- Registry team, Regional Directorate of Health of Casablanca, Morocco
| | - Hanane Bourezgui
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| | - Souha Sahraoui
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| | - Marilys Corbex
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Mhamed Harif
- Centre Hospitalier Mohammed VI, Marrakech, Morocco
| | - Abdellatif Benider
- Department of Radiotherapy-oncology, Ibn Rochd University Hospital, Casablanca, Hassan II University, Morocco
| |
Collapse
|
49
|
Abstract
Pancreatic cancer remains one of the most challenging of all cancers. Genetic risk factors are believed to play a major role, but other than genes coding for blood group, genetic risks for sporadic cases remain elusive. However, several germline mutations have been identified that lead to hereditary pancreatic cancer, familial pancreatic cancer, and increased risk for pancreatic cancer as part of a familial cancer syndrome. The most important genes with variants increasing risk for pancreatic cancer include BRCA1, BRCA2, PALB2, ATM, CDKN2A, APC, MLH1, MSH2, MSH6, PMS2, PRSS1, and STK11. Recognition of members of high-risk families is important for understanding pancreatic cancer biology, for recommending risk reduction strategies and, in some cases, initiating cancer surveillance programs. Because the best methods for surveillance have not been established, the recommendation to refer at-risk patients to centers with ongoing research programs in pancreatic cancer surveillance is supported.
Collapse
|
50
|
Abstract
This article reviews the genetics and incipient pathology of familial pancreatic cancer and the screening modalities in current use, and summarizes the outcomes of reported screening programs.
Collapse
Affiliation(s)
- Adam W Templeton
- Department of Gastroenterology, Digestive Diseases Center, University of Washington, Box Number 356424, 1959 Northeast Pacific Street, Seattle, WA 98195, USA.
| | | |
Collapse
|