1
|
Sharma R, Kumar S, Komal K, Ghosh R, Thakur S, Pal RR, Kumar M. Comprehensive insights into pancreatic cancer treatment approaches and cutting-edge nanocarrier solutions: from pathology to nanomedicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04094-y. [PMID: 40202672 DOI: 10.1007/s00210-025-04094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. It is characterized by poor prognosis, high mortality, and recurrence rates. Various modifiable and non-modifiable risk factors are associated with pancreatic cancer incidence. Available treatments for pancreatic cancer include surgery, chemotherapy, radiotherapy, photodynamic therapy, supportive care, targeted therapy, and immunotherapy. However, the survival rates for PC are very low. Regrettably, despite efforts to enhance prognosis, the survival rate of pancreatic cancer remains relatively low. Therefore, it is essential to investigate new approaches to improve pancreatic cancer treatment. By synthesizing current knowledge and identifying existing gaps, this article provides a comprehensive overview of risk factors, pathology, conventional treatments, targeted therapies, and recent advancements in nanocarriers for its treatment, along with various clinical trials and patents that justify the safety and efficacy of innovative carriers for drug delivery systems. Ultimately, this review underscores the potential of these innovative formulations to improve outcomes and contribute significantly to the advancement of Pancreatic Cancer treatment. Together, these insights highlight nano-formulations as a promising frontier for effectively treating Pancreatic Cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
2
|
Kimoto N, Miyashita Y, Yata Y, Aketa T, Yabumoto M, Sakata Y, Washio T, Takashima S, Kitakaze M. Metabolic syndrome is linked to most cancers incidence. Heart Vessels 2025; 40:350-360. [PMID: 39384596 DOI: 10.1007/s00380-024-02474-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024]
Abstract
Since many people die of either cancers or cardiovascular diseases worldwide, it is important to find the clinical pitfall that provokes cardiovascular diseases and cancer overall. Since metabolic syndrome (MetS) is largely linked to cardiovascular diseases, we have come to consider that MetS, even in its early state, may prime the occurrence of cancers overall. Indeed, the importance of MetS in causing pancreatic cancer has been proved using our large medical database. We analyzed Japanese healthcare and clinical data in 2005, who were followed up until 2020 and we examined the incidence of major cancers. At the enrollment, we examined the presence or absence of MetS judged by either Japanese criteria or NCEP/ATPIII. Of 2.7 million subjects without missing data, 102,930; 200,231; 237,420; 63,435; 76,172; and 2,422 subjects suffered lung, stomach, colon, liver and prostate cancer, respectively, and myelogenous leukemia during follow-up. MetS, defined by Japanese criteria, increased (p < 0.005 each) the incidence of cancer with a hazard ratio (HR) of 1.03-1.47 for lung, stomach, colon, liver, prostate cancers, and myelogenous leukemia. According to Japanese criteria, cancer incidence in the pre-stage MetS group was comparable to the MetS group. The results were almost identical when we defined MetS using NCEP ATP III. Taken together, we conclude that MetS is linked to majority of cancers.
Collapse
Affiliation(s)
- Naoki Kimoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- Non Profit Organization Think of Medicine in Science, 3-7-11, Minamisumiyoshi, Sumiyoshi-ku, Osaka, Japan
| | - Yohei Miyashita
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Yutaka Yata
- Hanwa Memorial Hospital, 3-5-8 Minamisumiyoshi, Sumiyoshi-ku, Osaka, Japan
| | - Takeshi Aketa
- ASCLEPIUS INC, 3-6-2 Minamisumiyoshi, Sumiyoshi-ku, Osaka, Japan
| | - Masami Yabumoto
- Hanwa Memorial Hospital, 3-5-8 Minamisumiyoshi, Sumiyoshi-ku, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Takashi Washio
- The Institute of Scientific and Industrial Research, Osaka University, 1-1 Yamadaoka, Suita, Osaka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
- The Osaka Medical Research Foundation for Intractable Diseases, 2-6-29 Abikohigashi, Sumiyoshi-ku, Osaka, Japan
| | - Masafumi Kitakaze
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Hanwa Memorial Hospital, 3-5-8 Minamisumiyoshi, Sumiyoshi-ku, Osaka, Japan.
- The Osaka Medical Research Foundation for Intractable Diseases, 2-6-29 Abikohigashi, Sumiyoshi-ku, Osaka, Japan.
| |
Collapse
|
3
|
Paranal RM, Wood LD, Klein AP, Roberts NJ. Understanding familial risk of pancreatic ductal adenocarcinoma. Fam Cancer 2024; 23:419-428. [PMID: 38609521 PMCID: PMC11660179 DOI: 10.1007/s10689-024-00383-2] [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: 01/27/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that is the result of an accumulation of sequential genetic alterations. These genetic alterations can either be inherited, such as pathogenic germline variants that are associated with an increased risk of cancer, or acquired, such as somatic mutations that occur during the lifetime of an individual. Understanding the genetic basis of inherited risk of PDAC is essential to advancing patient care and outcomes through improved clinical surveillance, early detection initiatives, and targeted therapies. In this review we discuss factors associated with an increased risk of PDAC, the prevalence of genetic variants associated with an increased risk in patients with PDAC, estimates of PDAC risk in carriers of pathogenic germline variants in genes associated with an increased risk of PDAC. The role of common variants in pancreatic cancer risk will also be discussed.
Collapse
Affiliation(s)
- Raymond M Paranal
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Human Genetics Predoctoral Training Program, the McKusick-Nathans Department of Genetic Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alison P Klein
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD, USA.
| | - Nicholas J Roberts
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Archasappawat S, Al-Musawi F, Liu P, Lee E, Hwang CI. Familial Pancreatic Cancer Research: Bridging Gaps in Basic Research and Clinical Application. Biomolecules 2024; 14:1381. [PMID: 39595558 PMCID: PMC11592027 DOI: 10.3390/biom14111381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Familial pancreatic cancer (FPC) represents a significant yet underexplored area in pancreatic cancer research. Basic research efforts are notably limited, and when present, they are predominantly centered on the BRCA1 and BRCA2 mutations due to the scarcity of other genetic variants associated with FPC, leading to a limited understanding of the broader genetic landscape of FPC. This review examines the current state of FPC research, focusing on the molecular mechanisms driving pancreatic ductal adenocarcinoma (PDAC) progression. It highlights the role of homologous recombination (HR) and its therapeutic exploitation via synthetic lethality with PARP inhibitors in BRCA1/2-deficient tumors. The review discusses various pre-clinical models of FPC, including conventional two-dimensional (2D) cell lines, patient-derived organoids (PDOs), patient-derived xenografts (PDXs), and genetically engineered mouse models (GEMMs), as well as new advancements in FPC research.
Collapse
Affiliation(s)
- Suyakarn Archasappawat
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| | - Fatimah Al-Musawi
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Peiyi Liu
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - EunJung Lee
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
| | - Chang-il Hwang
- Department of Microbiology and Molecular Genetics, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; (S.A.); (F.A.-M.); (P.L.)
- University of California Davis Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
5
|
Ahmed TM, Kawamoto S, Lopez-Ramirez F, Yasrab M, Hruban RH, Fishman EK, Chu LC. Early detection of pancreatic cancer in the era of precision medicine. Abdom Radiol (NY) 2024; 49:3559-3573. [PMID: 38761272 DOI: 10.1007/s00261-024-04358-w] [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: 03/31/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/20/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer-related mortality and it is often diagnosed at advanced stages due to non-specific clinical presentation. Disease detection at localized disease stage followed by surgical resection remains the only potentially curative treatment. In this era of precision medicine, a multifaceted approach to early detection of PDAC includes targeted screening in high-risk populations, serum biomarkers and "liquid biopsies", and artificial intelligence augmented tumor detection from radiologic examinations. In this review, we will review these emerging techniques in the early detection of PDAC.
Collapse
Affiliation(s)
- Taha M Ahmed
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Felipe Lopez-Ramirez
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Linda C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Hospital, Baltimore, MD, USA.
| |
Collapse
|
6
|
Palmstedt E, Månsson M, Kollberg KS, Carlsson S, Hellström M, Wallström J, Hugosson J, Arnsrud Godtman R. How a population-based cohort of men estimate lifetime risk of prostate cancer in a survey before entering a prostate cancer screening trial in Sweden? BMJ Open 2024; 14:e083562. [PMID: 39153780 PMCID: PMC11331866 DOI: 10.1136/bmjopen-2023-083562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/19/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVES Investigating men's perceived lifetime risk of prostate cancer. DESIGN Survey-based study to men invited for prostate-specific antigen (PSA) screening in the GÖTEBORG-2 trial between September 2015 and June 2020. SETTING 38 775 men in the Gothenburg area, Sweden, were invited for PSA-testing and participated in a survey. PARTICIPANTS 17 980 men participated in PSA-testing, of whom 13 189 completed the survey. In addition, 1264 men answered the survey only. INTERVENTIONS Before having the PSA-test, men answered an electronic survey and estimated their lifetime risk of receiving a prostate cancer diagnosis on a visual analogue scale from 0% to 100%. MAIN OUTCOME MEASURES The primary outcome was the median lifetime risk estimation, which was compared with Wilcoxon test to an anticipated lifetime risk of 20% (based on GÖTEBORG-1 trial). The secondary outcome was to determine factors associated with risk estimation in a multivariable linear regression model: previous prostate examination, family history, physical exercise, healthy diet, comorbidity, alcohol consumption, smoking, education level, marital status, urinary symptoms and erectile dysfunction. RESULTS Among PSA-tested men, the median estimated lifetime risk of prostate cancer was 30% (IQR 19% to 50%), corresponding to a 10 percentage-points higher estimation compared with the anticipated risk (p<0.001). Family history of prostate cancer, moderate to severe urinary symptoms and mild to moderate erectile dysfunction were associated with >5 percentage-points higher risk estimation. Similar results were obtained for non-PSA-tested men. CONCLUSIONS Most men overestimated their prostate cancer risk which underscores the importance of providing them accurate information about prostate cancer. TRIAL REGISTRATION NUMBER ISRCTN94604465.
Collapse
Affiliation(s)
- Emmeli Palmstedt
- Department of Urology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Marianne Månsson
- Department of Urology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Karin Stinesen Kollberg
- Department of Urology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
- Social Work, University of Gothenburg Faculty of Social Science, Gothenburg, Sweden
| | - Sigrid Carlsson
- Department of Surgery and Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York City, New York, USA
- Translational Medicine, Division of Urological Cancers, Lund University Medical Faculty, Lund, Sweden
| | - Mikael Hellström
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Radiology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Jonas Wallström
- Department of Radiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Radiology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
| | - Jonas Hugosson
- Department of Urology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
- Department of Urology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rebecka Arnsrud Godtman
- Department of Urology, University of Gothenburg Institute of Clinical Sciences, Goteborg, Sweden
- Department of Urology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
7
|
Limijadi EKS, Muniroh M, Prajoko YW, Tjandra KC, Respati DRP. The role of germline BRCA1 & BRCA2 mutations in familial pancreatic cancer: A systematic review and meta-analysis. PLoS One 2024; 19:e0299276. [PMID: 38809921 PMCID: PMC11135687 DOI: 10.1371/journal.pone.0299276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/15/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Familial Pancreatic Cancer (FPC) presents a notable risk, with 3-10% of pancreatic adenocarcinoma cases having a family history. Studies link FPC to syndromes like HBOC, suggesting BRCA1/BRCA2 mutations play a role. BRCA gene functions in DNA repair impact FPC management, influencing sensitivity to therapies like PARP inhibitors. Identifying mutations not only aids FPC treatment but also reveals broader cancer risks. However, challenges persist in selectively applying genetic testing due to cost constraints. This Systematic Review focuses on BRCA1/BRCA2 significance in FPC, diagnostic criteria, prognostic value, and limitations. METHOD Original articles published from 2013 to January 2023 were sourced from databases such as Scopus, PubMed, ProQuest, and ScienceDirect. Inclusion criteria comprised observational cohort or diagnostic studies related to the role of BRCA1/2 mutation in correlation to familial pancreatic cancer (FPC), while article reviews, narrative reviews, and non-relevant content were excluded. The assessment of bias used ROBINS-I, and the results were organized using PICOS criteria in a Google spreadsheet table. The systematic review adhered to the PRISMA 2020 checklist. RESULT We analyzed 9 diagnostic studies encompassing 1325 families and 4267 patients from Italy, USA, and Poland. Despite the limitation of limited homogenous PICO studies, our findings effectively present evidence. BRCA1/2 demonstrates benefits in detecting first-degree relatives FPC involvement with 2.26-10 times higher risk. These mutation findings also play an important role since with the BRCA1/2 targeted therapy, Poly-ADP Ribose Polymerase inhibitors (PARP) may give better outcomes of FPC treatment. Analysis of BRCA1 and BRCA2 administration's impact on odds ratio (OR) based on six and five studies respectively. BRCA1 exhibited non-significant effects (OR = 1.26, P = 0.51), while BRCA2 showed significance (OR = 1.68, P = 0.04). No heterogeneity observed, indicating consistent results. Further research on BRCA1 is warranted. CONCLUSION Detecting the BRCA1/2 mutation gene offers numerous advantages, particularly in its correlation with FPC. For diagnostic and prognostic purposes, testing is strongly recommended for first-degree relatives, who face a significantly higher risk (2.26-10 times) of being affected. Additionally, FPC patients with identified BRCA1/2 mutations exhibit a more favorable prognosis compared to the non-mutated population. This is attributed to the availability of targeted BRCA1/2 therapy, which maximizes treatment outcomes.
Collapse
Affiliation(s)
- Edward Kurnia Setiawan Limijadi
- Doctoral Study Program of Medical and Health Science, Universitas Diponegoro, Semarang, Indonesia
- Faculty of Medicine, Department of Clinical Pathology, Universitas Diopnegoro, Semarang, Indonesia
| | - Muflihatul Muniroh
- Faculty of Medicine, Department of Physiology, Universitas Diponegoro, Semarang, Indonesia
| | - Yan Wisnu Prajoko
- Faculty of Medicine, Department of Surgical Oncology, Universitas Diponegoro, Semarang, Indonesia
- Kariadi General Hospital, Semarang, Indonesia
| | - Kevin Christian Tjandra
- Kariadi General Hospital, Semarang, Indonesia
- Faculty of Medicine, Departement of Medicine, Universitas Diponegoro, Semarang, Indonesia
| | - Danendra Rakha Putra Respati
- Kariadi General Hospital, Semarang, Indonesia
- Faculty of Medicine, Departement of Medicine, Universitas Diponegoro, Semarang, Indonesia
| |
Collapse
|
8
|
Huang FF, Cui WH, Ma LY, Chen Q, Liu Y. Crosstalk of nervous and immune systems in pancreatic cancer. Front Cell Dev Biol 2023; 11:1309738. [PMID: 38099290 PMCID: PMC10720593 DOI: 10.3389/fcell.2023.1309738] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Pancreatic cancer is a highly malignant tumor known for its extremely low survival rate. The combination of genetic disorders within pancreatic cells and the tumor microenvironment contributes to the emergence and progression of this devastating disease. Extensive research has shed light on the nature of the microenvironmental cells surrounding the pancreatic cancer, including peripheral nerves and immune cells. Peripheral nerves release neuropeptides that directly target pancreatic cancer cells in a paracrine manner, while immune cells play a crucial role in eliminating cancer cells that have not evaded the immune response. Recent studies have revealed the intricate interplay between the nervous and immune systems in homeostatic condition as well as in cancer development. In this review, we aim to summarize the function of nerves in pancreatic cancer, emphasizing the significance to investigate the neural-immune crosstalk during the advancement of this malignant cancer.
Collapse
Affiliation(s)
- Fei-Fei Huang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Wen-Hui Cui
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Lan-Yue Ma
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Chen
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
9
|
Chen X, Meyer MA, Kemppainen JL, Horibe M, Chandra S, Majumder S, Petersen GM, Rabe KG. Risk of Syndrome-Associated Cancers Among First-Degree Relatives of Patients With Pancreatic Ductal Adenocarcinoma With Pathogenic or Likely Pathogenic Germline Variants. JAMA Oncol 2023; 9:955-961. [PMID: 37200008 PMCID: PMC10196930 DOI: 10.1001/jamaoncol.2023.0806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 02/06/2023] [Indexed: 05/19/2023]
Abstract
Importance Increased cancer risk in first-degree relatives of probands with pancreatic ductal adenocarcinoma (PDAC probands) who carry pathogenic or likely pathogenic germline variants (PGVs) in cancer syndrome-associated genes encourages cascade genetic testing. To date, unbiased risk estimates for the development of cancers on a gene-specific basis have not been assessed. Objective To quantify the risk of development of PDAC and extra-PDAC among first-degree relatives of PDAC probands who carry a PGV in 1 of 9 cancer syndrome-associated genes-ATM, BRCA1, BRCA2, PALB2, MLH1, MSH2, MSH6, PMS2, and CDKN2A. Design, Setting, and Participants This case series focused on first-degree relatives of PDAC probands carrying PGVs in specific cancer syndrome-associated genes. The cohort comprised clinic-ascertained patients enrolled in the Mayo Clinic Biospecimen Resource for Pancreas Research registry with germline genetic testing. In total, 234 PDAC probands carrying PGVs were drawn from the prospective research registry of 4562 participants who had undergone genetic testing of cancer syndrome-associated genes. Demographic and cancer-related family histories were obtained by questionnaire. The data were collected from October 1, 2000, to December 31, 2021. Main Outcomes and Measures For the PDAC probands, the genetic test results of the presence of PGVs in 9 cancer syndrome-associated genes were obtained by clinical testing. Cancers (ovary, breast, uterus or endometrial, colon, malignant melanoma, and pancreas) among first-degree relatives were reported by the probands. Standardized incidence ratios (SIRs) were used to estimate cancer risks among first-degree relatives of PDAC probands carrying a PGV. Results In total, 1670 first-degree relatives (mean [SD] age, 58.1 [17.8] years; 853 male [51.1%]) of 234 PDAC probands (mean [SD] age, 62.5 [10.1] years; 124 male [53.0%]; 219 [94.4%] White; 225 [98.7%] non-Hispanic or non-Latino]) were included in the study. There was a significantly increased risk of ovarian cancer in female first-degree relatives of probands who had variants in BRCA1 (SIR, 9.49; 95% CI, 3.06-22.14) and BRCA2 (SIR, 3.72; 95% CI, 1.36-8.11). Breast cancer risks were higher with BRCA2 variants (SIR, 2.62; 95% CI, 1.89-3.54). The risks of uterine or endometrial cancer (SIR, 6.53; 95% CI, 2.81-12.86) and colon cancer (SIR, 5.83; 95% CI, 3.70-8.75) were increased in first-degree relatives of probands who carried Lynch syndrome mismatch repair variants. Risk of PDAC was also increased for variants in ATM (SIR, 4.53; 95% CI, 2.69-7.16), BRCA2 (SIR, 3.45; 95% CI, 1.72-6.17), CDKN2A (SIR, 7.38; 95% CI, 3.18-14.54), and PALB2 (SIR, 5.39; 95% CI, 1.45-13.79). Melanoma risk was elevated for first-degree relatives of probands with CDKN2A variants (SIR, 7.47; 95% CI, 3.97-12.77). Conclusions and Relevance In this case series, the presence of PGVs in 9 cancer syndrome-associated genes in PDAC probands was found to be associated with increased risk of 6 types of cancers in first-degree relatives. These gene-specific PDAC and extra-PDAC cancer risks may provide justification for clinicians to counsel first-degree relatives about the relevance and importance of genetic cascade testing, with the goal of higher uptake of testing.
Collapse
Affiliation(s)
- Xuan Chen
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, Minnesota
| | - Margaret A Meyer
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis
| | | | - Masayasu Horibe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shruti Chandra
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Gloria M Petersen
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Kari G Rabe
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
10
|
Dite GS, Spaeth E, Wong CK, Murphy NM, Allman R. Predicting 10-Year Risk of Pancreatic Cancer Using a Combined Genetic and Clinical Model. GASTRO HEP ADVANCES 2023; 2:979-989. [PMID: 39130772 PMCID: PMC11308393 DOI: 10.1016/j.gastha.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/12/2023] [Indexed: 08/13/2024]
Abstract
Background and Aims Pancreatic cancer has the poorest 5-year survival rate of any major solid tumor, but when diagnosed at an early stage, survival rates improve. Population screening is impractical because pancreatic cancer is rare with a lifetime risk of 1.7%, but accurate risk stratification in the general population could enable health care providers to focus early detection strategies to at-risk individuals. Here, we validate a combined risk prediction model that integrates a polygenic risk score and a clinical risk model. Methods Using the UK Biobank, we conducted a prospective cohort study assessing 10-year pancreatic cancer risks based on a polygenic risk score, a clinical risk score, and a combined risk score. We assessed the association, discrimination, calibration, cumulative hazards, and standardized incidence ratios compared to population incidence rates for the risk scores. We also conducted net reclassification analyses. Results While all of the risk scores discriminated well between affected and unaffected participants, the combined risk score - with a Harrell's C-index of 0.714 (95% confidence interval [CI] = 0.698, 0.730) - discriminated better than both the polygenic risk score (P = .001) and the clinical risk score (P = .02). In terms of calibration, there was no problem with dispersion for the combined risk score (β = 0.952, 95% CI = 0.865-1.039, P = .3) and overall there was a small overestimation of risk (α = -0.089, 95% CI = -0.156 to -0.021, P = .009). Participants in the top decile of 10-year risk were at 1.413 (95% CI = 1.242-1.607) times population risk. Conclusion The combined risk score was able to identify individuals at substantially increased risk of pancreatic cancer and to whom targeted screening could be useful.
Collapse
Affiliation(s)
| | - Erika Spaeth
- Phenogen Sciences Inc, Charlotte, North Carolina
| | - Chi Kuen Wong
- Genetic Technologies Limited, Fitzroy, Victoria, Australia
| | | | - Richard Allman
- Genetic Technologies Limited, Fitzroy, Victoria, Australia
| |
Collapse
|
11
|
Copur MS, Tun SM, Vargas L, Merani S, Wedel W, Duckert R, Horn A, Lintel N, Herold D, Lavudi S. Unusual dMMR Phenotype Locally Advanced Pancreatic Ductal Adenocarcinoma with Germline and Somatic BRCA2 Mutation in a Jehovah Witness Patient. Clin Colorectal Cancer 2023; 22:160-165. [PMID: 36404245 DOI: 10.1016/j.clcc.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Mehmet Sitki Copur
- Mary Lanning Healthcare, Morrison Cancer Center, Hastings, NE; University of Nebraska Medical Center, Omaha, NE.
| | - Soe Min Tun
- Mary Lanning Healthcare, Morrison Cancer Center, Hastings, NE
| | | | | | | | - Randy Duckert
- Mary Lanning Healthcare, Morrison Cancer Center, Hastings, NE
| | - Adam Horn
- Mary Lanning Healthcare Pathology, Hastings, NE
| | | | | | - Swathi Lavudi
- Prairie Center Internal Medicine & Nephrology, Green Island, NE
| |
Collapse
|
12
|
Ha CSR, Müller-Nurasyid M, Petrera A, Hauck SM, Marini F, Bartsch DK, Slater EP, Strauch K. Proteomics biomarker discovery for individualized prevention of familial pancreatic cancer using statistical learning. PLoS One 2023; 18:e0280399. [PMID: 36701413 PMCID: PMC9879447 DOI: 10.1371/journal.pone.0280399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/28/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The low five-year survival rate of pancreatic ductal adenocarcinoma (PDAC) and the low diagnostic rate of early-stage PDAC via imaging highlight the need to discover novel biomarkers and improve the current screening procedures for early diagnosis. Familial pancreatic cancer (FPC) describes the cases of PDAC that are present in two or more individuals within a circle of first-degree relatives. Using innovative high-throughput proteomics, we were able to quantify the protein profiles of individuals at risk from FPC families in different potential pre-cancer stages. However, the high-dimensional proteomics data structure challenges the use of traditional statistical analysis tools. Hence, we applied advanced statistical learning methods to enhance the analysis and improve the results' interpretability. METHODS We applied model-based gradient boosting and adaptive lasso to deal with the small, unbalanced study design via simultaneous variable selection and model fitting. In addition, we used stability selection to identify a stable subset of selected biomarkers and, as a result, obtain even more interpretable results. In each step, we compared the performance of the different analytical pipelines and validated our approaches via simulation scenarios. RESULTS In the simulation study, model-based gradient boosting showed a more accurate prediction performance in the small, unbalanced, and high-dimensional datasets than adaptive lasso and could identify more relevant variables. Furthermore, using model-based gradient boosting, we discovered a subset of promising serum biomarkers that may potentially improve the current screening procedure of FPC. CONCLUSION Advanced statistical learning methods helped us overcome the shortcomings of an unbalanced study design in a valuable clinical dataset. The discovered serum biomarkers provide us with a clear direction for further investigations and more precise clinical hypotheses regarding the development of FPC and optimal strategies for its early detection.
Collapse
Affiliation(s)
- Chung Shing Rex Ha
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Medicine, Institute for Medical Information Processing, Chair of Genetic Epidemiology, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
- * E-mail:
| | - Martina Müller-Nurasyid
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Medicine, Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germanys
- Faculty of Medicine, Institute for Medical Information Processing, Pettenkofer School of Public Health Munich, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefanie M. Hauck
- Research Unit Protein Science and Metabolomics and Proteomics Core Facility, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Detlef K. Bartsch
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University, Marburg, Germany
| | - Emily P. Slater
- Department of Visceral-, Thoracic- and Vascular Surgery, Philipps University, Marburg, Germany
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Medicine, Institute for Medical Information Processing, Chair of Genetic Epidemiology, Biometry, and Epidemiology (IBE), LMU Munich, Munich, Germany
| |
Collapse
|
13
|
Porter N, Laheru D, Lau B, He J, Zheng L, Narang A, Roberts NJ, Canto MI, Lennon AM, Goggins MG, Hruban RH, Klein AP. Risk of Pancreatic Cancer in the Long-Term Prospective Follow-Up of Familial Pancreatic Cancer Kindreds. J Natl Cancer Inst 2022; 114:1681-1688. [PMID: 36029239 PMCID: PMC9745433 DOI: 10.1093/jnci/djac167] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/27/2022] [Accepted: 08/25/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND A family history of pancreatic cancer is associated with increased pancreatic cancer risk. However, risk estimates for individuals in kindreds with an aggregation of pancreatic cancer (>1 relative) are imprecise because of small samples sizes or potentially impacted by biases inherent in retrospective data. OBJECTIVE The objective of this study is to determine the age-specific pancreatic cancer risk as a function of family history using prospective data. METHODS We compared pancreatic cancer incidence (n = 167) in 21 141 individuals from 4433 families enrolled in the National Familial Pancreatic Cancer Registry with that expected based on Surveillance Epidemiology and End Results data and estimated the cumulative probability of pancreatic cancer using competing risk regression. RESULTS Familial pancreatic kindred members (kindreds with pancreatic cancer in 2 first-degree relatives [FDRs] or a pathogenic variant) had a standardized incidence ratio of 4.86 (95% confidence interval [CI] = 4.01 to 5.90), and sporadic kindred members (kindreds not meeting familial criteria) had a standardized incidence ratio of 2.55 (95% CI = 1.95 to 3.34). Risk in familial pancreatic cancer kindreds increased with an increasing number of FDRs with pancreatic cancer, with a standardized incidence ratio of 3.46 (95% CI = 2.52 to 4.76), 5.44 (95% CI = 4.07 to 7.26), and 10.78 (95% CI = 6.87 to 16.89) for 1, 2, and 3 or more FDRs with pancreatic cancer, respectively. Risk was also higher among individuals with a family history of young-onset (aged younger than 50 years) pancreatic cancer. CONCLUSION Pancreatic cancer risk is strongly dependent on family history, including both the degree of relationship(s) and age of onset of pancreatic cancer in relatives. These risk estimates will help inform the design of early detection studies and the risk and benefit analysis of screening trials.
Collapse
Affiliation(s)
- Nancy Porter
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Daniel Laheru
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Bryan Lau
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jin He
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amol Narang
- Division of Radiation Oncology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nicholas J Roberts
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Marcia I Canto
- Division of Gastroenterology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Anne Marie Lennon
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Radiation Oncology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Michael G Goggins
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
- Division of Gastroenterology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Alison P Klein
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
- Division of Gastroenterology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Lee CL, Holter S, Borgida A, Dodd A, Ramotar S, Grant R, Wasson K, Elimova E, Jang RW, Moore M, Kim TK, Khalili K, Moulton CA, Gallinger S, O’Kane GM, Knox JJ. Germline BRCA2 variants in advanced pancreatic acinar cell carcinoma: A case report and review of literature. World J Gastroenterol 2022; 28:6421-6432. [PMID: 36533108 PMCID: PMC9753052 DOI: 10.3748/wjg.v28.i45.6421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 11/02/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Pancreatic acinar cell carcinoma (PACC) is a rare tumor. Up to 45% of PACCs have alterations in the DNA damage repair pathway and 23% harbor rearrangements in the BRAF or RAF1 genes. We present a PACC case with a germline BRCA2 likely pathogenic variant (LPV) to highlight the impact of genomic testing on treatment decisions and patient outcomes. In our larger case series, we provide clinic-based information on additional 10 PACC patients treated in our center.
CASE SUMMARY A 70-year-old male was diagnosed with advanced PACC. At presentation, he was cachectic with severe arthralgia despite prednisolone and a skin rash that was later confirmed to be panniculitis. He was treated with modified FOLFIRINOX (mFFX) with the knowledge of the germline BRCA2 LPV. Following 11 cycles of mFFX, a computed tomography (CT) scan demonstrated significant tumor response in the pancreatic primary and hepatic metastases, totaling 70% from baseline as per Response Evaluation Criteria in Solid Tumors. Resolution of the skin panniculitis was also noted. We identified two additional PACCs with druggable targets in our case series. Our data contribute to practical evidence for the value of germline and somatic profiling in the management of rare diseases like PACC.
CONCLUSION This patient and others in our larger case series highlight the importance of genomic testing in PACC with potential utility in personalized treatment.
Collapse
Affiliation(s)
- Cha Len Lee
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Spring Holter
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Ayelet Borgida
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Anna Dodd
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Stephanie Ramotar
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Robert Grant
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Kristy Wasson
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Elena Elimova
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Raymond W Jang
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Malcolm Moore
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Tae Kyoung Kim
- Department of Medical Imaging, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Korosh Khalili
- Department of Medical Imaging, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Carol-Anne Moulton
- Hepatobiliary/Pancreatic Surgical Program, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Steven Gallinger
- Hepatobiliary/Pancreatic Surgical Program, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Grainne M O’Kane
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| | - Jennifer J Knox
- Division of Medical Oncology and Hematology, Wallace McCain Center for Pancreatic Cancer, Princess Margaret Cancer Centre, Toronto M5G1Z5, ON, Canada
| |
Collapse
|
15
|
Afghani E, Klein AP. Pancreatic Adenocarcinoma: Trends in Epidemiology, Risk Factors, and Outcomes. Hematol Oncol Clin North Am 2022; 36:879-895. [PMID: 36154788 PMCID: PMC10548451 DOI: 10.1016/j.hoc.2022.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pancreatic cancer is one of the most lethal cancers in the world; it is a silent disease in which symptoms do not present until advanced stages, thereby reducing the 5-year survival rate to 10%. The global burden of pancreatic cancer has doubled over the past 25 years despite advancements in medicine. This review aims to discuss the global trends and disparities in pancreatic cancer, as well as the up-to-date literature on the known risk factors. A better understanding of these risk factors will reduce mortality by providing opportunities to screen these patients as well as counseling on lifestyle modifications.
Collapse
Affiliation(s)
- Elham Afghani
- Johns Hopkins School of Medicine, 1830 E Monument Street, Room 436, Baltimore, MD 21205, USA
| | - Alison P Klein
- Johns Hopkins School of Medicine, 1830 E Monument Street, Room 436, Baltimore, MD 21205, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Johns Hopkins Bloomberg School of Public Health, 1550 Orleans Street, Baltimore, MD 21231, USA.
| |
Collapse
|
16
|
Vanek P, Urban O, Zoundjiekpon V, Falt P. Current Screening Strategies for Pancreatic Cancer. Biomedicines 2022; 10:biomedicines10092056. [PMID: 36140157 PMCID: PMC9495594 DOI: 10.3390/biomedicines10092056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a dreaded malignancy with a dismal 5-year survival rate despite maximal efforts on optimizing treatment strategies. Radical surgery is the only potential curative procedure. Unfortunately, the majority of patients are diagnosed with locally advanced or metastatic disease, which renders them ineligible for curative resection. Early detection of PDAC is thus considered to be the most effective way to improve survival. In this regard, pancreatic screening has been proposed to improve results by detecting asymptomatic stages of PDAC and its precursors. There is now evidence of benefits of systematic surveillance in high-risk individuals, and the current guidelines emphasize the potential of screening to affect overall survival in individuals with genetic susceptibility syndromes or familial occurrence of PDAC. Here we aim to summarize the current knowledge about screening strategies for PDAC, including the latest epidemiological data, risk factors, associated hereditary syndromes, available screening modalities, benefits, limitations, as well as management implications.
Collapse
|
17
|
Panthangi V, Cyril Kurupp AR, Raju A, Luthra G, Shahbaz M, Almatooq H, Foucambert P, Esbrand FD, Zafar S, Khan S. Association Between Helicobacter pylori Infection and the Risk of Pancreatic Cancer: A Systematic Review Based on Observational Studies. Cureus 2022; 14:e28543. [PMID: 36185865 PMCID: PMC9518818 DOI: 10.7759/cureus.28543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/29/2022] [Indexed: 12/24/2022] Open
Abstract
Helicobacter pylori (H. pylori) bacterial infection has long been scrutinized as one of the potential risk factors for the development of pancreatic cancer with quite inconsistent and unequivocal data. Little is known about the risk factors involved with this malignancy. In this systematic review, we aimed to examine the relationship between H. pylori infection and pancreatic cancer based on the evidence from the existing observational studies across the world. We searched major electronic databases such as PubMed, MEDLINE, Science Direct, and Cochrane Library. After a careful and thorough screening process, we selected 15 observation studies for this systematic review. Six of 15 studies found a significant association between H. pylori infection and pancreatic cancer. Additionally, four of these studies found a significant relationship between the cytotoxin-associated gene A strain of H. pylori and pancreatic cancer. Based on the evidence from the selected studies, a weak association was observed between H. pylori infection and cancer of the pancreas, especially in European and Asian populations compared to the North American population. The cross-sectional evidence from the case-control studies only suggests the existence of an association but does not provide substantial evidence of the causative relationship. Further large-scale, prospective cohort studies are warranted in the future to understand this contradictory relationship better.
Collapse
Affiliation(s)
- Venkatesh Panthangi
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Anjumol Raju
- Pediatrics, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Gaurav Luthra
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mahrukh Shahbaz
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Halah Almatooq
- Dermatology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Paul Foucambert
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Faith D Esbrand
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sana Zafar
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
18
|
Association Between Family History and Risk of Pancreatic Cancer in Patients With BRCA1 and BRCA2 Pathogenic Variants. Pancreas 2022; 51:733-738. [PMID: 36395396 DOI: 10.1097/mpa.0000000000002104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVES Current guidelines limit pancreatic cancer screening to those BRCA1/2 patients who have a family history of pancreatic cancer. We aimed to assess the association between family history and risk of pancreatic neoplasms in BRCA1/2 patients. METHODS We reviewed medical records of BRCA1/2 patients followed at our institution between 1995 and 2020. Family history was defined as those with a first-degree relative with pancreatic cancer. We compared the incidence and prevalence of pancreatic neoplasms between patients with and without family history of pancreatic cancer. RESULTS We identified 56 BRCA1/2 patients with family history and 238 without family history of pancreatic cancer. No difference between these groups was noted in age, race, or sex. Mean follow-up interval for BRCA1/2 patients was 4.6 years (range, 0-19.7 years). There was no significant difference in prevalence (19.6% vs 12.6; P = 0.3) or incidence (29% vs 14.1%; P = 0.08) of branch-duct intraductal papillary mucinous neoplasm between the 2 groups. No association between family history and pancreatic cancer risk was noted. Only 1 of 10 BRCA1/2 patients with pancreatic cancer had a family history. CONCLUSIONS Our results do not support using family history to determine eligibility for pancreatic cancer screening.
Collapse
|
19
|
Overbeek KA, Levink IJM, Koopmann BDM, Harinck F, Konings ICAW, Ausems MGEM, Wagner A, Fockens P, van Eijck CH, Groot Koerkamp B, Busch ORC, Besselink MG, Bastiaansen BAJ, van Driel LMJW, Erler NS, Vleggaar FP, Poley JW, Cahen DL, van Hooft JE, Bruno MJ. Long-term yield of pancreatic cancer surveillance in high-risk individuals. Gut 2022; 71:1152-1160. [PMID: 33820756 PMCID: PMC9120399 DOI: 10.1136/gutjnl-2020-323611] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We aimed to determine the long-term yield of pancreatic cancer surveillance in hereditary predisposed high-risk individuals. DESIGN From 2006 to 2019, we prospectively enrolled asymptomatic individuals with an estimated 10% or greater lifetime risk of pancreatic ductal adenocarcinoma (PDAC) after obligatory evaluation by a clinical geneticist and genetic testing, and subjected them to annual surveillance with both endoscopic ultrasonography (EUS) and MRI/cholangiopancreatography (MRI/MRCP) at each visit. RESULTS 366 individuals (201 mutation-negative familial pancreatic cancer (FPC) kindreds and 165 PDAC susceptibility gene mutation carriers; mean age 54 years, SD 9.9) were followed for 63 months on average (SD 43.2). Ten individuals developed PDAC, of which four presented with a symptomatic interval carcinoma and six underwent resection. The cumulative PDAC incidence was 9.3% in the mutation carriers and 0% in the FPC kindreds (p<0.001). Median PDAC survival was 18 months (range 1-32). Surgery was performed in 17 individuals (4.6%), whose pathology revealed 6 PDACs (3 T1N0M0), 7 low-grade precursor lesions, 2 neuroendocrine tumours <2 cm, 1 autoimmune pancreatitis and in 1 individual no abnormality. There was no surgery-related mortality. EUS detected more solid lesions than MRI/MRCP (100% vs 22%, p<0.001), but less cystic lesions (42% vs 83%, p<0.001). CONCLUSION The diagnostic yield of PDAC was substantial in established high-risk mutation carriers, but non-existent in the mutation-negative proven FPC kindreds. Nevertheless, timely identification of resectable lesions proved challenging despite the concurrent use of two imaging modalities, with EUS outperforming MRI/MRCP. Overall, surveillance by imaging yields suboptimal results with a clear need for more sensitive diagnostic markers, including biomarkers.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Iris J M Levink
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Brechtje D M Koopmann
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Femme Harinck
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ingrid C A W Konings
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Margreet G E M Ausems
- Division Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paul Fockens
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Casper H van Eijck
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Olivier R C Busch
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Barbara A J Bastiaansen
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lydi M J W van Driel
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Nicole S Erler
- Department of Biostatistics, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Frank P Vleggaar
- Department of Gastroenterology & Hepatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan-Werner Poley
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeanin E van Hooft
- Department of Gastroenterology & Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marco J Bruno
- Department of Gastroenterology & Hepatology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
20
|
Calderwood AH, Sawhney MS, Thosani NC, Rebbeck TR, Wani S, Canto MI, Fishman DS, Golan T, Hidalgo M, Kwon RS, Riegert-Johnson DL, Sahani DV, Stoffel EM, Vollmer CM, Al-Haddad MA, Amateau SK, Buxbaum JL, DiMaio CJ, Fujii-Lau LL, Jamil LH, Jue TL, Law JK, Lee JK, Naveed M, Pawa S, Storm AC, Qumseya BJ. American Society for Gastrointestinal Endoscopy guideline on screening for pancreatic cancer in individuals with genetic susceptibility: methodology and review of evidence. Gastrointest Endosc 2022; 95:827-854.e3. [PMID: 35183359 DOI: 10.1016/j.gie.2021.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 02/08/2023]
Affiliation(s)
- Audrey H Calderwood
- Section of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Mandeep S Sawhney
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nirav C Thosani
- Center for Interventional Gastroenterology at UTHealth, McGovern Medical School, Houston, Texas, USA
| | - Timothy R Rebbeck
- Harvard TH Chan School of Public Health and Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sachin Wani
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Marcia I Canto
- Division of Gastroenterology and Hepatology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Douglas S Fishman
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Talia Golan
- Cancer Center, Sheba Medical Center, Yehuda, Israel
| | - Manuel Hidalgo
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Richard S Kwon
- Division of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas L Riegert-Johnson
- Department of Clinical Genomics and Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Dushyant V Sahani
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Elena M Stoffel
- Division of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles M Vollmer
- Department of Surgery, Penn Medicine, Philadelphia, Pennsylvania, USA
| | - Mohammad A Al-Haddad
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stuart K Amateau
- Division of Gastroenterology Hepatology and Nutrition, University of Minnesota Medical Center, Minneapolis, Minnesota, USA
| | - James L Buxbaum
- Division of Gastrointestinal and Liver Diseases, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Christopher J DiMaio
- Department of Gastroenterology, Mount Sinai School of Medicine, New York, New York, USA
| | - Larissa L Fujii-Lau
- Department of Gastroenterology, The Queen's Medical Center, Honolulu, Hawaii, USA
| | - Laith H Jamil
- Section of Gastroenterology and Hepatology, Beaumont Health, Royal Oak, Michigan, and Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
| | - Terry L Jue
- Department of Gastroenterology, The Permanente Medical Group, San Francisco, California, USA
| | - Joanna K Law
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Virginia Mason Medical Center, Seattle, Washington, USA
| | - Jeffrey K Lee
- Department of Gastroenterology, Kaiser Permanente San Francisco Medical Center, San Francisco, California, USA
| | - Mariam Naveed
- Advent Health Medical Group, Gastroenterology/Hepatology, Advent Health Hospital Altamonte Springs, Altamonte Springs, Florida, USA
| | - Swati Pawa
- Department of Gastroenterology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Andrew C Storm
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Bashar J Qumseya
- Department of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
21
|
Rabe KG, Stevens MA, Hernández AT, Chandra S, Hubbard JM, Kemppainen JL, Majumder S, Petersen GM. Pancreatic cancer risk to siblings of probands in bilineal cancer settings. Genet Med 2022; 24:1008-1016. [PMID: 35227607 PMCID: PMC9326771 DOI: 10.1016/j.gim.2022.01.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Pancreatic cancer (PC) risk is increased in families, but PC risk and risk perception have been understudied when both parents have cancer. METHODS An unbiased method defining cancer triads (proband with PC and both parents with cancer) in a prospective registry estimated risk of PC to probands' siblings in triad group 1 (no parent with PC), group 2 (1 parent with PC), and group 3 (both parents with PC). We estimated standardized incidence ratios (SIRs) using a Surveillance, Epidemiology, and End Results (SEER) reference. We also estimated the risk when triad probands carried germline pathogenic/likely pathogenic variants in any of the 6 PC-associated genes (ATM, BRCA1, BRCA2, CDKN2A, MLH1, and TP53). PC risk perception/concern was surveyed in siblings and controls. RESULTS Risk of PC was higher (SIR = 3.5; 95% CI = 2.2-5.2) in 933 at-risk siblings from 297 triads. Risk increased by triad group: 2.8 (95% CI = 1.5-4.5); 4.5 (95% CI = 1.6-9.7); and 21.2 (95% CI = 4.3-62.0). SIR in variant-negative triads was 3.0 (95% CI = 1.6-5.0), whereas SIR in variant-positive triads was 10.0 (95% CI = 3.2-23.4). Siblings' perceived risk/concern of developing PC increased by triad group. CONCLUSION Sibling risks were 2.8- to 21.2-fold higher than that of the general population. Positive variant status increased the risk in triads. Increasing number of PC cases in a triad was associated with increased concern and perceived PC risk.
Collapse
Affiliation(s)
- Kari G Rabe
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Maria A Stevens
- Division of Health Care Policy and Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Amanda Toledo Hernández
- School of Medicine, Medical Science Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Shruti Chandra
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | | | | | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Gloria M Petersen
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN.
| |
Collapse
|
22
|
Sawhney MS, Calderwood AH, Thosani NC, Rebbeck TR, Wani S, Canto MI, Fishman DS, Golan T, Hidalgo M, Kwon RS, Riegert-Johnson DL, Sahani DV, Stoffel EM, Vollmer CM, Qumseya BJ. ASGE guideline on screening for pancreatic cancer in individuals with genetic susceptibility: summary and recommendations. Gastrointest Endosc 2022; 95:817-826. [PMID: 35183358 DOI: 10.1016/j.gie.2021.12.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Mandeep S Sawhney
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Audrey H Calderwood
- Section of Gastroenterology and Hepatology, Dartmouth-Hitchcock Medical Center, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Nirav C Thosani
- Center for Interventional Gastroenterology at UT Health, McGovern Medical School, Houston, Texas, USA
| | - Timothy R Rebbeck
- Harvard TH Chan School of Public Health and Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Sachin Wani
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Marcia I Canto
- Division of Gastroenterology and Hepatology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Douglas S Fishman
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Manuel Hidalgo
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, New York, USA
| | - Richard S Kwon
- Division of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas L Riegert-Johnson
- Department of Clinical Genomics and Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Dushyant V Sahani
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Elena M Stoffel
- Division of Gastroenterology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles M Vollmer
- Department of Surgery, Penn Medicine, Philadelphia, Pennsylvania, USA
| | - Bashar J Qumseya
- Department of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
23
|
Hijioka S, Morizane C, Takaori K, Okusaka T. Study protocol for a multi-institutional prospective surveillance study among kindreds with familial pancreatic cancer and individuals with hereditary pancreatic cancer syndrome: The Diamond Study. Pancreatology 2022; 22:534-538. [PMID: 35443912 DOI: 10.1016/j.pan.2022.04.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Family history of pancreatic cancer (PC) and some hereditary cancer syndromes are risk factors for PC. Previous studies suggest that conducting surveillance for kindreds at high risk for familial PC may be useful for diagnoses at the stage where resections can still be implemented; however, there is insufficient evidence linking surveillance and increased rates of resectable PC. METHODS We launched a surveillance study for kindreds with familial PC and individuals with hereditary PC syndrome, titled the "Diamond Study," in June 2020. This Japanese national multi-institutional prospective intervention study has been initiated to conduct evaluations within a prospective clinical trial format. RESULTS The primary endpoint is the fraction of patients with resectable PC among patients with PC found through surveillance interventions. Endoscopic ultrasound and magnetic resonance imaging combined with magnetic resonance cholangiopancreatography will be performed alternatively every 6 months for up to 15 years, with 400 as the predicted number of registered participants and a predicted registration period of 10 years. CONCLUSION We intend to scientifically prove the usefulness of surveillance for kindreds with familial PC and individuals with hereditary PC syndrome to improve PC prognoses.
Collapse
Affiliation(s)
- Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
24
|
Zhang AMY, Chu KH, Daly BF, Ruiter T, Dou Y, Yang JCC, de Winter TJJ, Chhuor J, Wang S, Flibotte S, Zhao YB, Hu X, Li H, Rideout EJ, Schaeffer DF, Johnson JD, Kopp JL. Effects of hyperinsulinemia on pancreatic cancer development and the immune microenvironment revealed through single-cell transcriptomics. Cancer Metab 2022; 10:5. [PMID: 35189981 PMCID: PMC8862319 DOI: 10.1186/s40170-022-00282-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Hyperinsulinemia is independently associated with increased risk and mortality of pancreatic cancer. We recently reported that genetically reduced insulin production resulted in ~ 50% suppression of pancreatic intraepithelial neoplasia (PanIN) precancerous lesions in mice. However, only female mice remained normoglycemic, and only the gene dosage of the rodent-specific Ins1 alleles was tested in our previous model. Moreover, we did not delve into the molecular and cellular mechanisms associated with modulating hyperinsulinemia. METHODS We studied how reduced Ins2 gene dosage affects PanIN lesion development in both male and female Ptf1aCreER;KrasLSL-G12D mice lacking the rodent-specific Ins1 gene (Ins1-/-). We generated control mice having two alleles of the wild-type Ins2 gene (Ptf1aCreER;KrasLSL-G12D;Ins1-/-;Ins2+/+) and experimental mice having one allele of Ins2 gene (Ptf1aCreER;KrasLSL-G12D;Ins1-/-;Ins2+/-). We then performed thorough histopathological analyses and single-cell transcriptomics for both genotypes and sexes. RESULTS High-fat diet-induced hyperinsulinemia was transiently or modestly reduced in female and male mice, respectively, with only one allele of Ins2. This occurred without dramatically affecting glucose tolerance. Genetic reduction of insulin production resulted in mice with a tendency for less PanIN and acinar-to-ductal metaplasia (ADM) lesions. Using single-cell transcriptomics, we found hyperinsulinemia affected multiple cell types in the pancreas, with the most statistically significant effects on local immune cell types that were highly represented in our sampled cell population. Specifically, hyperinsulinemia modulated pathways associated with protein translation, MAPK-ERK signaling, and PI3K-AKT signaling, which were changed in epithelial cells and subsets of immune cells. CONCLUSIONS These data suggest a potential role for the immune microenvironment in hyperinsulinemia-driven PanIN development. Together with our previous work, we propose that mild suppression of insulin levels may be useful in preventing pancreatic cancer by acting on multiple cell types.
Collapse
Affiliation(s)
- Anni M Y Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Ken H Chu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Brian F Daly
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Titine Ruiter
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Yan Dou
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Jenny C C Yang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Twan J J de Winter
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Justin Chhuor
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Su Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Stephane Flibotte
- Life Sciences Institute Bioinformatics Core Facility, University of British Columbia, Vancouver, Canada
| | - Yiwei Bernie Zhao
- Biomedical Research Centre, School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Xiaoke Hu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Hong Li
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - David F Schaeffer
- Department of Pathology and Laboratory and Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada.
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
25
|
Sagami R, Sato T, Mizukami K, Motomura M, Okamoto K, Fukuchi S, Otsuka Y, Abe T, Ono H, Mori K, Wada K, Iwaki T, Nishikiori H, Honda K, Amano Y, Murakami K. Diagnostic Strategy of Early Stage Pancreatic Cancer via Clinical Predictor Assessment: Clinical Indicators, Risk Factors and Imaging Findings. Diagnostics (Basel) 2022; 12:diagnostics12020377. [PMID: 35204468 PMCID: PMC8871200 DOI: 10.3390/diagnostics12020377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Early detection of pancreatic ductal adenocarcinoma (PDAC) in the general population is difficult due to unknown clinical characteristics. This study was conducted to clarify the factors associated with early stage PDAC. Well-known symptoms and factors associated with PDAC were classified into clinical indicators, risk factors, and imaging findings concomitant with early stage PDAC. To analyze these factors for the detection of patients with early stage PDAC compared to patients without PDAC, we constructed new diagnostic strategies. The factors of 35 patients with early stage PDAC (stage 0 and IA) and 801 patients without PDAC were compared retrospectively. Clinical indicators; presence and number of indicators, elevated pancreatic enzyme level, tumor biomarker level, acute pancreatitis history, risk factors; familial pancreatic cancer, diabetes mellitus, smoking history, imaging findings; presence and number of findings, and main pancreatic duct dilation were significant factors for early stage PDAC detection. A new screening strategy to select patients who should be examined by imaging modalities from evaluating clinical indicators and risk factors and approaching a definitive diagnosis by evaluating imaging findings had a relatively high sensitivity, specificity, and areas under the curve of 80.0%, 80.8%, and 0.80, respectively. Diagnosis based on the new category and strategy may be reasonable for early stage PDAC detection.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita 870-1151, Japan; (R.S.); (T.S.); (H.N.)
| | - Takao Sato
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita 870-1151, Japan; (R.S.); (T.S.); (H.N.)
| | - Kazuhiro Mizukami
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
- Correspondence: ; Tel.: +81-97-586-6193
| | - Mitsuteru Motomura
- Department of Gastroenterology, Oita Red Cross Hospital, 3-2-37 Chiyo-Machi, Oita 870-0033, Japan;
| | - Kazuhisa Okamoto
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| | - Satoshi Fukuchi
- Department of Gastroenterology, Oita City Medical Association Almeida Memorial Hospital, 1509-2 Miyazaki, Oita 870-1195, Japan; (S.F.); (K.W.)
| | - Yuichiro Otsuka
- Department of Gastroenterology, Oita Medical Center, 2-11-45 Yokota, Oita 870-0263, Japan;
| | - Takashi Abe
- Department of Gastroenterology, Oita Kouseiren Tsurumi Hospital, 4333 Tsurumi, Beppu 874-8585, Japan;
| | - Hideki Ono
- Department of Gastroenterology, Oita Prefectural Hospital, 2-8-1 Bunyo, Oita 870-8511, Japan;
| | - Kei Mori
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| | - Kurato Wada
- Department of Gastroenterology, Oita City Medical Association Almeida Memorial Hospital, 1509-2 Miyazaki, Oita 870-1195, Japan; (S.F.); (K.W.)
| | - Tomoyuki Iwaki
- Department of Endoscopy, Urawa Kyosai Hospital, 3-15-31 Harayama, Saitama 336-0931, Japan; (T.I.); (Y.A.)
| | - Hidefumi Nishikiori
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita 870-1151, Japan; (R.S.); (T.S.); (H.N.)
| | - Koichi Honda
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| | - Yuji Amano
- Department of Endoscopy, Urawa Kyosai Hospital, 3-15-31 Harayama, Saitama 336-0931, Japan; (T.I.); (Y.A.)
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu 879-5593, Japan; (K.O.); (K.M.); (K.H.); (K.M.)
| |
Collapse
|
26
|
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
|
27
|
Kasuga A, Okamoto T, Udagawa S, Mori C, Mie T, Furukawa T, Yamada Y, Takeda T, Matsuyama M, Sasaki T, Ozaka M, Ueki A, Sasahira N. Molecular Features and Clinical Management of Hereditary Pancreatic Cancer Syndromes and Familial Pancreatic Cancer. Int J Mol Sci 2022; 23:1205. [PMID: 35163129 PMCID: PMC8835700 DOI: 10.3390/ijms23031205] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/17/2022] Open
Abstract
Hereditary pancreatic cancers are caused by several inherited genes. Familial pancreatic cancer is defined as pancreatic cancer arising in a patient with at least two first-degree relatives with pancreatic cancer in the absence of an identified genetic cause. Hereditary pancreatic cancer syndromes and familial pancreatic cancers account for about 10% of pancreatic cancer cases. Germline mutations in BRCA1, BRCA2, ATM, PALB2, CDKN2A, STK11, and TP53 and mismatch repair genes (MLH1, MSH2, MSH6, PMS2, and EPCAM) are among the well-known inherited susceptibility genes. Currently available targeted medications include poly (ADP-ribose) polymerase inhibitors (PARP) for cases with mutant BRCA and immune checkpoint inhibitors for cases with mismatch repair deficiency. Loss of heterozygosity of hereditary pancreatic cancer susceptibility genes such as BRCA1/2 plays a key role in carcinogenesis and sensitivity to PARP inhibitors. Signature 3 identified by whole genome sequencing is also associated with homologous recombination deficiency and sensitivity to targeted therapies. In this review, we summarize molecular features and treatments of hereditary pancreatic cancer syndromes and surveillance procedures for unaffected high-risk cases. We also review transgenic murine models to gain a better understanding of carcinogenesis in hereditary pancreatic cancer.
Collapse
Affiliation(s)
- Akiyoshi Kasuga
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takeshi Okamoto
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Shohei Udagawa
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan;
| | - Chinatsu Mori
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takafumi Mie
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Yuto Yamada
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Tsuyoshi Takeda
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Masato Matsuyama
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Takashi Sasaki
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| | - Arisa Ueki
- Department of Clinical Genetics, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan;
| | - Naoki Sasahira
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; (T.O.); (C.M.); (T.M.); (T.F.); (Y.Y.); (T.T.); (M.M.); (T.S.); (M.O.); (N.S.)
| |
Collapse
|
28
|
Momayez Sanat Z, Masoudi S, Mansouri M, Ghamarzad Shishavan N, Jameshorani M, Pourshams A. Diabetes Mellitus, Obesity, and Risk of Pancreatic Ductal Adenocarcinoma: a Large Case-Control Study from Iran. Middle East J Dig Dis 2021; 13:15-20. [PMID: 34712433 PMCID: PMC8531941 DOI: 10.34172/mejdd.2021.198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/11/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a deadly, globally increasing cancer. The causes of PC are still insufficiently known, however smoking, diabetes mellitus (DM), and obesity have been identified as risk factors of PC, mostly in the developed countries. We evaluated these risk factors and their contribution to PC among an Iranian population. METHODS Cases and controls were selected from patients who were registered to a tertiary gastrointestinal diseases referral hospital in Tehran, Iran, from Jan 2012 to Jan 2018. Information on risk factors was collected by personal interview using a structured questionnaire. Logistic regression models were used to calculate adjusted odds ratios (AORs) and 95% confidence intervals (CIs). RESULTS We recruited 470 new patients with histopathological PC diagnosis and 526 sex and age-matched controls. Cigarette-smoking [AOR: 1.65 (1.15-2.38)], opium use [AOR: 1.58 (1.06-2.35)], DM [AOR: 1.99 (1.31-3.02)], and having a history of any cancer in a first-degree family member [AOR: 1.53 (1.14-2.05)] were associated with an increased risk of PC. We did not find an association between obesity [AOR: 0.99 (0.71-1.38)] and PC. Approximately 4.6%, 5.9%, 8.2%, and 10.9% risk of PC were related to cigarette-smoking, opium use, DM, and family history of any cancer, respectively. CONCLUSION This study supports that DM is associated with PC risk; however, similar to many studies in Asia, obesity is not associated with PC in Iranians. DM has the highest impact on PC development in Iranian women.
Collapse
Affiliation(s)
- Zahra Momayez Sanat
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Masoudi
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Mansouri
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Ghamarzad Shishavan
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Jameshorani
- Department of Internal Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Akram Pourshams
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Digestive Oncology Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.,Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
29
|
Ranganath R, Chu Q. Global trends in pancreas cancer among Asia-Pacific population. J Gastrointest Oncol 2021; 12:S374-S386. [PMID: 34422401 DOI: 10.21037/jgo-20-118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
The incidence of pancreatic cancer is rising. Understanding trends of pancreatic cancer is crucial prior to putting policies and interventions in place. Countries with a high human development index (HDI) have a higher incidence, prevalence and mortality due to pancreatic cancer. This global trend is replicated in the Asia-Pacific countries with high HDI having higher incidence, prevalence and mortality due to pancreatic cancer. The incidence of pancreatic cancer is rising in the Asia-Pacific population as life expectancy increases with a rising HDI. Lack of good cancer registries has resulted in under reporting of pancreatic cancer in developing countries in the Asia-Pacific region. The mortality still remains high as in the Western world as most pancreatic cancers are diagnosed in an advanced stage of the disease due to non-availability of cost-effective screening tools with few patients receiving definitive care. Smoking, alcohol consumption, poor diet and obesity are significant modifiable risk factors contributing to the development of pancreatic cancer. Population based screening for pancreatic cancer is not cost-effective. Identification of hereditary and genetic factors in the Asia-Pacific population can help in targeted screening of high-risk individuals. Policies and interventions aimed at primary prevention have the greatest potential to be cost-effective yet impactful and reduce the disease burden.
Collapse
Affiliation(s)
- Rohit Ranganath
- Department of Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Quyen Chu
- Department of Surgery, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
30
|
Murali K, Dwarte TM, Nikfarjam M, Tucker KM, Vaughan RB, Efthymiou M, Collins A, Spigelman AD, Salmon L, Johns AL, Williams DB, Delatycki MB, John T, Stoita A. Significant detection of new germline pathogenic variants in Australian Pancreatic Cancer Screening Program participants. Hered Cancer Clin Pract 2021; 19:33. [PMID: 34399810 PMCID: PMC8365963 DOI: 10.1186/s13053-021-00190-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/09/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The Australian Pancreatic Cancer Screening Program (APCSP) offers endoscopic ultrasound surveillance for individuals at increased risk of pancreatic ductal adenocarcinoma (PDAC) with all participants requiring assessment by a Familial Cancer Service before or after study enrolment. METHODS Individuals aged 40-80 years (or 10 years younger than the earliest PDAC diagnosis) were eligible for APCSP study entry if they had 1) ≥ two blood relatives with PDAC (at least one of first-degree association); 2) a clinical or genetic diagnosis of Hereditary Pancreatitis or Peutz-Jeghers syndrome irrespective of PDAC family history; or 3) a known PDAC predisposition germline pathogenic variant (BRCA2, PALB2, CDKN2A, or Lynch syndrome) with ≥one PDAC-affected first- or second-degree relative. Retrospective medical record review was conducted for APCSP participants enrolled at the participating Australian hospitals from January 2011 to December 2019. We audited the genetic investigations offered by multiple Familial Cancer Services who assessed APCSP participants according to national guidelines, local clinical protocol and/or the availability of external research-funded testing, and the subsequent findings. Descriptive statistical analysis was performed using Microsoft Excel. RESULTS Of 189 kindreds (285 participants), 50 kindreds (71 participants) had a known germline pathogenic variant at enrolment (BRCA2 n = 35, PALB2 n = 6, CDKN2A n = 3, STK11 n = 3, PRSS1 n = 2, MLH1 n = 1). Forty-eight of 136 (35%) kindreds with no known germline pathogenic variant were offered mutation analysis; 89% was clinic-funded, with increasing self-funded testing since 2016. The relatively low rates of genetic testing performed reflects initial strict criteria for clinic-funded genetic testing. New germline pathogenic variants were detected in five kindreds (10.4%) after study enrolment (BRCA2 n = 3 kindreds, PALB2 n = 1, CDKN2A n = 1). Of note, only eight kindreds were reassessed by a Familial Cancer Service since enrolment, with a further 21 kindreds identified as being suitable for reassessment. CONCLUSION Germline pathogenic variants associated with PDAC were seen in 29.1% of our high-risk cohort (55/189 kindreds; 82/285 participants). Importantly, 10.4% of kindreds offered genetic testing were newly identified as having germline pathogenic variants, with majority being BRCA2. As genetic testing standards evolve rapidly in PDAC, 5-yearly reassessment of high-risk individuals by Familial Cancer Services is warranted.
Collapse
Affiliation(s)
- Krithika Murali
- Department of Clinical Genetics, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Tanya M Dwarte
- Australian Pancreatic Cancer Genome Initiative, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- Hereditary Cancer Centre, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
- Department of Gastroenterology, St Vincent's Hospital, Darlinghurst, NSW, 2010, Australia
| | - Mehrdad Nikfarjam
- Division of Surgery, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Katherine M Tucker
- Hereditary Cancer Centre, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
- University of New South Wales, St Vincent's Clinical School and Prince of Wales Clinical School, Randwick, NSW, 2031, Australia
| | - Rhys B Vaughan
- Department of Gastroenterology, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Marios Efthymiou
- Department of Gastroenterology, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Allison Collins
- Clinical Trials Unit, Olivia Newton John Cancer and Wellness Centre, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Allan D Spigelman
- University of New South Wales, St Vincent's Clinical School and Prince of Wales Clinical School, Randwick, NSW, 2031, Australia
- Cancer Genetics Unit, The Kinghorn Cancer Centre, St Vincent's Hospital, Darlinghurst, NSW, 2010, Australia
| | - Lucinda Salmon
- Department of Clinical Genetics, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Amber L Johns
- Australian Pancreatic Cancer Genome Initiative, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - David B Williams
- Department of Gastroenterology, St Vincent's Hospital, Darlinghurst, NSW, 2010, Australia
| | - Martin B Delatycki
- Department of Clinical Genetics, Austin Health, Heidelberg, VIC, 3084, Australia
| | - Thomas John
- Peter MacCallum Cancer Centre, Parkville, VIC, 3000, Australia
| | - Alina Stoita
- Department of Gastroenterology, St Vincent's Hospital, Darlinghurst, NSW, 2010, Australia.
- University of New South Wales, St Vincent's Clinical School and Prince of Wales Clinical School, Randwick, NSW, 2031, Australia.
| |
Collapse
|
31
|
Tan M, Brusgaard K, Gerdes AM, Mortensen MB, Detlefsen S, Schaffalitzky de Muckadell OB, Joergensen MT. Cohort profile and heritability assessment of familial pancreatic cancer: a nation-wide study. Scand J Gastroenterol 2021; 56:965-971. [PMID: 34165379 DOI: 10.1080/00365521.2021.1937697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Familial Pancreatic Cancer (FPC) is responsible for up to 10% of all cases of pancreatic ductal adenocarcinoma (PDAC). Individuals predisposed for FPC have an estimated lifetime risk of 16-39% of developing PDAC. While heritability of PDAC has been estimated to be 36% in a Nordic twin study, no heritability estimate specific on FPC has been reported. METHODS A national cohort of Danish families with predisposition for FPC is currently included in a screening program for PDAC at Odense University Hospital. Family members included in the screening program were interviewed for pedigree data including: cases of PDAC among first-degree relatives (FDRs) and number of affected/unaffected siblings. Heritability for FPC in the predisposed families was assessed by doubling the estimated intra-class correlation coefficient (ICC) from a random intercept logistic model fitted to data on FDRs. RESULTS Among families with predisposition for FPC, 83 cases of PDAC were identified. The median age at diagnosis of PDAC was 66 years, and median time from diagnosis to death was 7.6 months. A total of 359 individuals were found as unaffected FDRs of the 83 PDAC cases. The retrieved FDRs included a total of 247 individuals in sibship and 317 individuals in parent-offspring relatedness. We estimated an ICC of 0.25, corresponding to a narrow sense additive heritability estimate of 0.51 in the FPC family cohort. CONCLUSION We have established a nation-wide cohort of FPC families to facilitate clinical and genetic studies on FPC. The estimated heritability of 51% prominently underlines a strong genetic background of FPC.
Collapse
Affiliation(s)
- Ming Tan
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| | - Klaus Brusgaard
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Michael Bau Mortensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Ove B Schaffalitzky de Muckadell
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| | - Maiken Thyregod Joergensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| |
Collapse
|
32
|
Tan M, Brusgaard K, Gerdes AM, Mortensen MB, Detlefsen S, Schaffalitzky de Muckadell OB, Joergensen MT. Whole genome sequencing identifies rare germline variants enriched in cancer related genes in first degree relatives of familial pancreatic cancer patients. Clin Genet 2021; 100:551-562. [PMID: 34313325 PMCID: PMC9291090 DOI: 10.1111/cge.14038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/20/2022]
Abstract
First-degree relatives (FDRs) of familial pancreatic cancer (FPC) patients have increased risk of developing pancreatic ductal adenocarcinoma (PDAC). Investigating and understanding the genetic basis for PDAC susceptibility in FPC predisposed families may contribute toward future risk-assessment and management of high-risk individuals. Using a Danish cohort of 27 FPC families, we performed whole-genome sequencing of 61 FDRs of FPC patients focusing on rare genetic variants that may contribute to familial aggregation of PDAC. Statistical analysis was performed using the gnomAD database as external controls. Through analysis of heterozygous premature truncating variants (PTV), we identified cancer-related genes and cancer-driver genes harboring multiple germline mutations. Association analysis detected 20 significant genes with false discovery rate, q < 0.05 including: PALD1, LRP1B, COL4A2, CYLC2, ZFYVE9, BRD3, AHDC1, etc. Functional annotation showed that the significant genes were enriched by gene clusters encoding for extracellular matrix and associated proteins. PTV genes were over-represented by functions related to transport of small molecules, innate immune system, ion channel transport, and stimuli-sensing channels. In conclusion, FDRs of FPC patients carry rare germline variants related to cancer pathogenesis that may contribute to increased susceptibility to PDAC. The identified variants may potentially be useful for risk prediction of high-risk individuals in predisposed families.
Collapse
Affiliation(s)
- Ming Tan
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| | - Klaus Brusgaard
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Michael Bau Mortensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark.,Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Ove B Schaffalitzky de Muckadell
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| | - Maiken Thyregod Joergensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Medical Gastroenterology, Odense University Hospital, Odense, Denmark.,Odense Pancreas Center (OPAC), Odense University Hospital, Odense, Denmark
| |
Collapse
|
33
|
Cai J, Chen H, Lu M, Zhang Y, Lu B, You L, Zhang T, Dai M, Zhao Y. Advances in the epidemiology of pancreatic cancer: Trends, risk factors, screening, and prognosis. Cancer Lett 2021; 520:1-11. [PMID: 34216688 DOI: 10.1016/j.canlet.2021.06.027] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a malignancy with poor prognosis and high mortality. The recent increase in pancreatic cancer incidence and mortality has resulted in an increased number of studies on its epidemiology. This comprehensive and systematic literature review summarizes the advances in the epidemiology of pancreatic cancer, including its epidemiological trends, risk factors, risk prediction models, screening modalities, and prognosis. The risk factors for pancreatic cancers can be categorized as those related to individual characteristics, lifestyle and environment, and disease status. Several prediction models for pancreatic cancer have been developed in populations with new-onset diabetes or a family history of pancreatic cancer; however, these models require further validation. Despite recent progress in pancreatic cancer screening, the quantity and quality of related studies are also unsatisfactory, especially with respect to the identification of high-risk populations and development of effective screening modality. Apart from the populations with familial genetic risk and those at a high risk of sporadic pancreatic cancer, risk factors such as new-onset diabetes may be a new direction for timely intervention. We hope this work will provide new ideas for further prevention and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jie Cai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongda Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Ming Lu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Yuhan Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Bin Lu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Min Dai
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
34
|
Shih CY, Chattopadhyay A, Wu CH, Tien YW, Lu TP. Transcript annotation tool (TransAT): an R package for retrieving annotations for transcript-specific genetic variants. BMC Bioinformatics 2021; 22:350. [PMID: 34182919 PMCID: PMC8240296 DOI: 10.1186/s12859-021-04243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An individual's genetics play a role in how RNA transcripts are generated from DNA and consequently in their translation into protein. Transcriptional and translational profiling of patients furnishes the information that a specific marker is present; however, it fails to provide evidence whether the marker correlates with response to a therapeutic agent. A comparative analysis of the frequency of genetic variants, such as single nucleotide polymorphisms (SNPs), in diseased and general populations can identify pathogenic variants in individual patients. This is in part because SNPs have considerable effects on protein function and gene expression when they occur in coding regions and regulatory sequences, respectively. Therefore, a tool that can help users to obtain the allele frequency for a corresponding transcript is the need of the day. Several annotation tools such as SNPnexus and VariED are publicly available; however, none of them can use transcript IDs as input and provide the corresponding genomic positions of variants. RESULTS In this study, we developed an R package, called transcript annotation tool (TransAT), that provides (i) SNP ID and genomic position for a user-provided transcript ID from patients, and (ii) allele frequencies for the SNPs from publicly available global populations. All data elements are extracted, collected, and displayed in an easily downloadable format in two simple command lines. TransAT is available on Windows/Linux/MacOS and is operative for R version 4.0.4 or later. It is available at https://github.com/ShihChingYu/TransAT and can be downloaded and installed using devtools::install_github("ShihChingYu/TransAT", force=T) on the R execution page. Thereafter, all functions can be executed by loading the package into R with library(TransAT). CONCLUSIONS TransAT is a novel tool that seamlessly provides genetic annotations for queried transcripts. Such easily obtainable information would be greatly advantageous for physicians, assisting them to make individualized decisions about specific drug treatments. Moreover, allele frequencies from user-chosen global ethnic populations will highlight the importance of ethnicity and its effect on patient pathogenicity.
Collapse
Affiliation(s)
- Ching-Yu Shih
- Bioinformatics and Biostatistics Core, Centre of Genomic and Precision Medicine, National Taiwan University, Taipei, 10055, Taiwan
| | - Amrita Chattopadhyay
- Bioinformatics and Biostatistics Core, Centre of Genomic and Precision Medicine, National Taiwan University, Taipei, 10055, Taiwan
| | - Chien-Hui Wu
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, 10055, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzu-Pin Lu
- Bioinformatics and Biostatistics Core, Centre of Genomic and Precision Medicine, National Taiwan University, Taipei, 10055, Taiwan.
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, 10055, Taiwan.
| |
Collapse
|
35
|
Khalaf N, El-Serag HB, Abrams HR, Thrift AP. Burden of Pancreatic Cancer: From Epidemiology to Practice. Clin Gastroenterol Hepatol 2021; 19:876-884. [PMID: 32147593 PMCID: PMC8559554 DOI: 10.1016/j.cgh.2020.02.054] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/05/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide with 432,242 related deaths in 2018. Unlike other cancers, the incidence of pancreatic cancer continues to increase, with little improvement in survival rates. We review the epidemiologic features of pancreatic cancer, covering surveillance and early detection in high-risk persons. We summarize data on worldwide incidence and mortality and analyze the 1975-2016 data from 9 registries of the National Cancer Institute's Surveillance, Epidemiology, and End Results study, on the overall burden of pancreatic cancer as well as age-, sex-, and race-specific incidence, survival rates and trends. It is important to increase our knowledge of the worldwide and regional epidemiologic features of and risk factors for pancreatic cancer, to identify new approaches for prevention, surveillance, and treatment.
Collapse
Affiliation(s)
- Natalia Khalaf
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas.
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Hannah R Abrams
- Department of Internal Medicine, Baylor College of Medicine, Houston, Texas
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
36
|
Zhang AM, Wellberg EA, Kopp JL, Johnson JD. Hyperinsulinemia in Obesity, Inflammation, and Cancer. Diabetes Metab J 2021; 45:285-311. [PMID: 33775061 PMCID: PMC8164941 DOI: 10.4093/dmj.2020.0250] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
The relative insufficiency of insulin secretion and/or insulin action causes diabetes. However, obesity and type 2 diabetes mellitus can be associated with an absolute increase in circulating insulin, a state known as hyperinsulinemia. Studies are beginning to elucidate the cause-effect relationships between hyperinsulinemia and numerous consequences of metabolic dysfunctions. Here, we review recent evidence demonstrating that hyperinsulinemia may play a role in inflammation, aging and development of cancers. In this review, we will focus on the consequences and mechanisms of excess insulin production and action, placing recent findings that have challenged dogma in the context of the existing body of literature. Where relevant, we elaborate on the role of specific signal transduction components in the actions of insulin and consequences of chronic hyperinsulinemia. By discussing the involvement of hyperinsulinemia in various metabolic and other chronic diseases, we may identify more effective therapeutics or lifestyle interventions for preventing or treating obesity, diabetes and cancer. We also seek to identify pertinent questions that are ripe for future investigation.
Collapse
Affiliation(s)
- Anni M.Y. Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth A. Wellberg
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| | - Janel L. Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
37
|
Sipra QUAR, Islam M, Riaz IB, Zhaohui J, Babiker HM, Bekaii-Saab TS, Sonbol MB. Contemporary Management of Pancreatic Cancer from an Internist Perspective. Am J Med 2021; 134:576-586. [PMID: 33316248 DOI: 10.1016/j.amjmed.2020.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/23/2022]
Abstract
Primary care physicians are in a favorable position to curb the growing burden of pancreatic ductal adenocarcinoma. This review aims to provide an overview of pancreatic ductal adenocarcinoma from a primary care perspective, with a specific focus on risk factors, selection of high-risk individuals for screening, patient presentation at the primary-care clinic, and the role of the internist in supportive care. Overall, the internist is an essential member of the multidisciplinary care team with respect to optimizing patients' quality of life across various stages of the pancreatic cancer.
Collapse
Affiliation(s)
| | - Mahnoor Islam
- Dow Medical University, Karachi City, Sindh, Pakistan
| | | | | | | | | | | |
Collapse
|
38
|
Partelli S, Sclafani F, Barbu ST, Beishon M, Bonomo P, Braz G, de Braud F, Brunner T, Cavestro GM, Crul M, Trill MD, Ferollà P, Herrmann K, Karamitopoulou E, Neuzillet C, Orsi F, Seppänen H, Torchio M, Valenti D, Zamboni G, Zins M, Costa A, Poortmans P. European Cancer Organisation Essential Requirements for Quality Cancer Care (ERQCC): Pancreatic Cancer. Cancer Treat Rev 2021; 99:102208. [PMID: 34238640 DOI: 10.1016/j.ctrv.2021.102208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022]
Abstract
European Cancer Organisation Essential Requirements for Quality Cancer Care (ERQCC) are written by experts representing all disciplines involved in cancer care in Europe. They give patients, health professionals, managers and policymakers a guide to essential care throughout the patient journey. Pancreatic cancer is an increasing cause of cancer mortality and has wide variation in treatment and care in Europe. It is a major healthcare burden and has complex diagnosis and treatment challenges. Care must be carried out only in pancreatic cancer units or centres that have a core multidisciplinary team (MDT) and an extended team of health professionals detailed here. Such units are far from universal in European countries. To meet European aspirations for comprehensive cancer control, healthcare organisations must consider the requirements in this paper, paying particular attention to multidisciplinarity and patient-centred pathways from diagnosis, to treatment, to survivorship.
Collapse
Affiliation(s)
- Stefano Partelli
- European Society of Surgical Oncology (ESSO); IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Sclafani
- European Organisation for Research and Treatment of Cancer (EORTC); Institut Jules Bordet, Brussels, Belgium
| | - Sorin Traian Barbu
- Pancreatic Cancer Europe (PCE); Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Marc Beishon
- Cancer World, European School of Oncology (ESO), Milan, Italy
| | - Pierluigi Bonomo
- Flims Alumni Club (FAC); Careggi University Hospital, Florence, Italy
| | - Graça Braz
- European Oncology Nursing Society (EONS); Portuguese Oncology Institute, Porto, Portugal
| | - Filippo de Braud
- Organisation of European Cancer Institutes (OECI); IRCCS Foundation National Cancer Institute of Milan, Milan, Italy
| | - Thomas Brunner
- European Society for Radiotherapy and Oncology (ESTRO); Otto von Guericke University, Magdeburg, Germany
| | - Giulia Martina Cavestro
- European Hereditary Tumour Group (EHTG); IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mirjam Crul
- European Society of Oncology Pharmacy (ESOP); Amsterdam University Medical Centre, Netherlands
| | - Maria Die Trill
- International Psycho-Oncology Society (IPOS); ATRIUM: Psycho-Oncology & Clinical Psychology, Madrid, Spain
| | - Piero Ferollà
- International Neuroendocrine Cancer Alliance (INCA); Umbria Regional Cancer Network, Perugia, Italy
| | - Ken Herrmann
- European Association of Nuclear Medicine (EANM); University Hospital Essen, Essen, Germany
| | - Eva Karamitopoulou
- European Society of Pathology (ESP); Institute of Pathology, University of Bern, Bern, Switzerland
| | - Cindy Neuzillet
- International Society of Geriatric Oncology (SIOG), Institut Curie, Saint-Cloud, France
| | - Franco Orsi
- Cardiovascular and Interventional Radiological Society of Europe (CIRSE); European Institute of Oncology, Milan, Italy
| | - Hanna Seppänen
- Association of European Cancer Leagues (ECL); Helsinki University Hospital, Helsinki, Finland
| | - Martina Torchio
- Organisation of European Cancer Institutes (OECI); IRCCS Foundation National Cancer Institute of Milan, Milan, Italy
| | - Danila Valenti
- European Association for Palliative Care (EAPC); Palliative Care Network, AUSL Bologna, Bologna, Italy
| | - Giulia Zamboni
- European Society of Oncologic Imaging (ESOI); University Hospital Verona, Verona, Italy
| | - Marc Zins
- European Society of Radiology (ESR); Groupe hospitalier Paris Saint-Joseph, Paris, France
| | | | - Philip Poortmans
- European Cancer Organisation (ECCO); Iridium Kankernetwerk and University of Antwerp, Wilrijk-Antwerp, Belgium
| |
Collapse
|
39
|
Genkinger JM, Su GH, Santella RM. Identifying Novel Genetic Markers Through a Transcription-Wide Association Study: Can This Be a Path to Reducing the Burden of Pancreatic Cancer? J Natl Cancer Inst 2021; 112:977-978. [PMID: 31917439 DOI: 10.1093/jnci/djz247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/03/2020] [Indexed: 12/20/2022] Open
Affiliation(s)
- Jeanine M Genkinger
- Department of Epidemiology, Mailman School of Public Health, 722 w 168th St, Rm 712, Columbia University, New York, NY 10032, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA
| | - Gloria H Su
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, 1130 St. Nicolas Avenue, ICRC 10-04, New York, NY 10032, USA.,Department of Otolaryngology and Head and Neck Surgery, Columbia University Medical Center, 180 Fort Washington Avenue, 7th floor, New York, NY 10032, USA
| | - Regina M Santella
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, 1130 St Nicholas Ave, New York, NY 10032, USA.,Department of Environmental Health Sciences, Mailman School of Public Health, 630 West 168th St Room 16-410, Columbia University, New York, NY 10032, USA
| |
Collapse
|
40
|
Edelman HE, McClymont SA, Tucker TR, Pineda S, Beer RL, McCallion AS, Parsons MJ. SOX9 modulates cancer biomarker and cilia genes in pancreatic cancer. Hum Mol Genet 2021; 30:485-499. [PMID: 33693707 DOI: 10.1093/hmg/ddab064] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/02/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive form of cancer with high mortality. The cellular origins of PDAC are largely unknown; however, ductal cells, especially centroacinar cells (CACs), have several characteristics in common with PDAC, such as expression of SOX9 and components of the Notch-signaling pathway. Mutations in KRAS and alterations to Notch signaling are common in PDAC, and both these pathways regulate the transcription factor SOX9. To identify genes regulated by SOX9, we performed siRNA knockdown of SOX9 followed by RNA-seq in PANC-1s, a human PDAC cell line. We report 93 differentially expressed (DE) genes, with convergence on alterations to Notch-signaling pathways and ciliogenesis. These results point to SOX9 and Notch activity being in a positive feedback loop and SOX9 regulating cilia production in PDAC. We additionally performed ChIP-seq in PANC-1s to identify direct targets of SOX9 binding and integrated these results with our DE gene list. Nine of the top 10 downregulated genes have evidence of direct SOX9 binding at their promoter regions. One of these targets was the cancer stem cell marker EpCAM. Using whole-mount in situ hybridization to detect epcam transcript in zebrafish larvae, we demonstrated that epcam is a CAC marker and that Sox9 regulation of epcam expression is conserved in zebrafish. Additionally, we generated an epcam null mutant and observed pronounced defects in ciliogenesis during development. Our results provide a link between SOX9, EpCAM and ciliary repression that can be exploited in improving our understanding of the cellular origins and mechanisms of PDAC.
Collapse
Affiliation(s)
- Hannah E Edelman
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Sarah A McClymont
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Tori R Tucker
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, CA 92697, USA
| | - Santiago Pineda
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, CA 92697, USA
| | - Rebecca L Beer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Andrew S McCallion
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA
| | - Michael J Parsons
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 N. Broadway, 470 Miller Research Building, Baltimore, MD 21205, USA.,Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, CA 92697, USA
| |
Collapse
|
41
|
Abstract
Screening for pancreatic cancer (PC) in high-risk groups aimed to detect early cancers is currently done only in the research setting, and data on psychological outcomes of screening in these populations is scarce. To determine the psychological impact of a national Australian pancreatic screening program, a prospective study was conducted using validated psychological measures: impact of events scale (IES), psychological consequences questionnaire (PCQ) and the cancer worry scale. Measures were administered at baseline, 1-month and at 1-year post-enrolment and correlations with abnormal endoscopic ultrasound (EUS) results were calculated. Over a 6-year period, 102 participants were recruited to the screening program. Thirty-nine patients (38.2%) had an abnormal endoscopic ultrasound, and two patients (2.0%) were diagnosed with PC and two with other malignancies. Those with a personal history of cancer or a positive BRCA2 mutation demonstrated significantly increased worry about developing other types of cancer at baseline (p < 0.01). Irrespective of EUS result, there was a significant decrease of total IES score at 1 year (Z = - 2.0, p = 0.041). In patients with abnormal EUS results, there was a decrease in the total IES score at 1 year (Z = - 2.5, p = 0.011). In participants deemed to be most distressed at baseline based on their negative PCQ score, there was a significant decrease of the total PCQ (Z = - 3.2, p = 0.001), emotional (Z = - 3.0, p = 0.001), social (Z = 3.0, p = 0.001) and physical (Z = - 2.8, p = 0.002) subscale at 1-year post-intervention. This study provides evidence of the long-term psychological benefits of PC screening in high-risk patients. There was no negative impact of screening in the short-term and the positive benefits appeared at 1-year post-intervention irrespective of screening result.
Collapse
|
42
|
Van Tran T, Van Dao T, Nguyen KD, van Ta T, Vu KT, Trinh SH, Nguyen HC, Bui OT, Nguyen QT, Vu HD, Nguyen HL, Tran HT. Risk factors of Pancreatic Cancer in Vietnam: A Matched Case-Control Hospital-Based Study. Cancer Control 2021; 28:1073274821989320. [PMID: 33517745 PMCID: PMC8482704 DOI: 10.1177/1073274821989320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Data about the risk factors and pancreatic cancer in developing countries remain limited. We investigated for the first time the role of a number of risk factors (family cancer history, smoking, alcohol consumption, diabetes, inflammation disease, HBV infection) associated with pancreatic cancer among Vietnamese patients. Methods: We included all patients hospitalized at 4 Northern Vietnamese hospitals (Vietnam National Cancer Hospital, Bach Mai, Viet Duc, Thai Nguyen) and diagnosed with pancreatic cancer during the period from 2017 to 2019. Risk factors of eligible patients were collected and assessed the associations using a matched control study and logistic regression model analysis. Results: We identified 196 patients with diagnosis of pancreatic cancer of which 114 males and 82 females. The average age of the patient at the time of diagnosis was 58.28 years (standard deviation of 12.94, ranging from 25 to 87). Most of patients were diagnosed at advanced stage (85%). Smoking, diabetes, inflammation disease significantly increased the cancer risks (OR and 95% CI were 2.42 (1.38-4.37), 3.09 (1.54-6.68), 2.21 (1.42-3.45), respectively). HBV infection demonstrated a significant link with pancreatic cancer in univariate model (OR = 2.94 (1.08-9.36)), but not in multivariate model. However, cancer family history and alcohol drinkers did not show any significantly increased risk related to pancreatic cancer. Conclusions: Our finding showed smoking, diabetes, inflammation disease significantly increased the risk of pancreatic cancer in Vietnam.
Collapse
Affiliation(s)
- Thuan Van Tran
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,106156Hanoi Medical University, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - Tu Van Dao
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - Khac-Dung Nguyen
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - To van Ta
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,106156Hanoi Medical University, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | | | | | | | - Oanh Thi Bui
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - Quang Tien Nguyen
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - Hoan Duc Vu
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - Hiep Le Nguyen
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| | - Huong Thanh Tran
- Vietnam National Cancer Hospital, Hanoi, Vietnam.,106156Hanoi Medical University, Hanoi, Vietnam.,Vietnam National Cancer Institute, Hanoi, Vietnam
| |
Collapse
|
43
|
Rainone M, Singh I, Salo-Mullen EE, Stadler ZK, O'Reilly EM. An Emerging Paradigm for Germline Testing in Pancreatic Ductal Adenocarcinoma and Immediate Implications for Clinical Practice: A Review. JAMA Oncol 2021; 6:764-771. [PMID: 32053139 DOI: 10.1001/jamaoncol.2019.5963] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Importance Pancreatic ductal adenocarcinoma (PDAC) is a malignant neoplasm with a rising incidence and is a leading public health challenge. Pancreatic ductal adenocarcinoma has been well characterized genomically, with findings of therapeutic actionability that have substantive implications for clinical practice based on recent high-level evidence. Observations Pathogenic germline alterations (PGAs) are relatively common in individuals with PDAC, as evidenced in multiple recent data sets, with a frequency of approximately 10%. The most common PGAs are in BRCA1, BRCA2, and ATM and more rarely in PALB2, MLH1, MSH2, MSH6, PMS2, CDKN2A, and TP53, among others, with an aggregate frequency of 3.8% to 9.7%. These PGAs are of key interest owing to therapeutic actionability and the downstream identification of at-risk family members and possible hereditary cancer syndromes. Approximately 3% to 7% of individuals with PDAC harbor a BRCA1 or BRCA2 mutation, which are among the most frequently mutated genes in PDAC. Recent updates to the American Society of Clinical Oncology and the National Comprehensive Cancer Network guidelines recommend risk assessment for all individuals with PDAC irrespective of personal or family history or ethnicity. Treatment implications include the use of checkpoint inhibitor therapy for mismatch repair-deficient PDAC and the validation of poly-ADP (adenosine diphosphate)-ribose polymerase inhibitor (PARPi) therapy as a maintenance strategy in platinum-sensitive PDAC. Conclusions and Relevance With increasing evidence and slow improvement of outcomes, PDAC has entered the era of precision medicine. Germline mutations have been identified in key genes with an aggregate frequency of 3.8% to 9.7%, several of which are therapeutically actionable with platinum, PARPi, and checkpoint inhibitor therapy. Potential therapeutic targets need to be actively sought and identified.
Collapse
Affiliation(s)
- Michael Rainone
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Mount Sinai St Luke's and Mount Sinai West Hospitals, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Isha Singh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Mount Sinai St Luke's and Mount Sinai West Hospitals, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erin E Salo-Mullen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
44
|
Morani AC, Hanafy AK, Ramani NS, Katabathina VS, Yedururi S, Dasyam AK, Prasad SR. Hereditary and Sporadic Pancreatic Ductal Adenocarcinoma: Current Update on Genetics and Imaging. Radiol Imaging Cancer 2020; 2:e190020. [PMID: 33778702 DOI: 10.1148/rycan.2020190020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 08/08/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a genetically heterogeneous, biologically aggressive malignancy with a uniformly poor prognosis. While most pancreatic cancers arise sporadically, a small subset of PDACs develop in patients with hereditary and familial predisposition. Detailed studies of the rare hereditary syndromes have led to identification of specific genetic abnormalities that contribute to malignancy. For example, germline mutations involving BRCA1, BRCA2, PRSS1, and mismatch repair genes predispose patients to PDAC. While patients with Lynch syndrome develop a rare "medullary" variant of adenocarcinoma, intraductal papillary mucinous tumors are observed in patients with McCune-Albright syndrome. It is now well established that PDACs originate via a multistep progression from microscopic and macroscopic precursors due to cumulative genetic abnormalities. Improved knowledge of tumor genetics and oncologic pathways has contributed to a better understanding of tumor biology with attendant implications on diagnosis, management, and prognosis. In this article, the genetic landscape of PDAC and its precursors will be described, the hereditary syndromes that predispose to PDAC will be reviewed, and the current role of imaging in screening and staging assessment, as well as the potential role of molecular tumor-targeted imaging for evaluation of patients with PDAC and its precursors, will be discussed. Keywords: Abdomen/GI, Genetic Defects, Oncology, Pancreas Supplemental material is available for this article. © RSNA, 2020.
Collapse
Affiliation(s)
- Ajaykumar C Morani
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| | - Abdelrahman K Hanafy
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| | - Nisha S Ramani
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| | - Venkata S Katabathina
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| | - Sireesha Yedururi
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| | - Anil K Dasyam
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| | - Srinivasa R Prasad
- Departments of Diagnostic Radiology (A.C.M., A.K.H., S.Y., S.R.P.) and Pathology (N.S.R.), The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1473, Houston, TX 77030-4009; Department of Radiology, University of Texas at San Antonio, San Antonio, Tex (V.S.K.); and Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pa (A.K.D.)
| |
Collapse
|
45
|
Abstract
Pancreatic cancer (PC) is an increasingly common disease worldwide. Having a better understanding of worldwide and regional epidemiologic features and risk factors of PC is essential to identify new approaches for prevention, early diagnosis, surveillance, and treatment. In this article, we review the epidemiologic features and risk factors for PC and discuss opportunities and challenges of PC future treatment.
Collapse
Affiliation(s)
- Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jinxin Tao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
46
|
Abstract
Despite extensive research in the pathogenesis, early detection, and therapeutic approaches of pancreatic ductal adenocarcinoma (PDAC), it remains a devastating and incurable disease. As the global incidence and prevalence of PDAC continue to rise, there is a pressing need to place strong emphasis on its prevention. Although it is widely recognized that cigarette smoking, a potentially modifiable risk factor, has been linked to PDAC development, its contribution to prognosis is still uncertain. Moreover, the mechanistic pathways of PDAC progression secondary to smoking are various and lack a summative narration. Herein, we update and summarize the direct and indirect roles cigarette smoking plays on PDAC development, review literature to conclude the impact cigarette smoking has on prognosis, and postulate a comprehensive mechanism for cigarette smoking-induced PDAC.
Collapse
|
47
|
Kohli DR, Smith KR, Wong J, Yu Z, Boucher K, Faigel DO, Pannala R, Burt RW, Curtin K, Samadder NJ. Familial pancreatic cancer risk: a population-based study in Utah. J Gastroenterol 2019; 54:1106-1112. [PMID: 31240435 DOI: 10.1007/s00535-019-01597-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/03/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Pancreas adenocarcinoma (PC) has an undefined hereditary component. We quantified the familial risk of PC among relatives of patients diagnosed with PC and stratified it based on anatomic location of PC and age and sex of the proband. METHODS This is a retrospective, population-based, case-control study of PC diagnosed in Utah between 1980 and 2011. The Utah population database and cancer registry were used to identify index patients with PC. The risk of PC in first-degree relatives (FDRs), second-degree relatives (SDRs), and first cousins (FCs) of probands was compared with randomly selected sex- and age-matched population controls. RESULTS A total of 4,095 patients and 40,933 controls were identified. The relative risk (RR) of PC was 1.76 (95% CI 1.35-2.29) in FDRs, 1.42 (95% CI 1.18-1.7) in SDRs and 1.08 (95% CI 0.95-1.23) in FCs of probands compared to relatives of PC-free controls. The RR were elevated in FDRs (1.96, 95% CI 1.45-2.65), SDRs (1.54, 95% CI 1.19-1.98) and FCs (1.18, 95% CI 1.0-1.64) of female probands. Among probands diagnosed as < 65 years, RR was 2.12 (95% CI 1.37-3.28) in FDRs, 1.94 (95% CI 1.44-2.62) in SDRs, and 1.28 (95% CI 1.0-1.64) in FCs. Overall, the RR for PC was elevated in FDRs regardless of the anatomic location of PC. DISCUSSION There is an increased risk of PC in FDR and more distant relatives of patients with PC. Relatives of female patients with PC and patients diagnosed at age < 65 years are at a significantly increased risk of PC.
Collapse
Affiliation(s)
- Divyanshoo R Kohli
- Division of Gastroenterology and Hepatology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA.,Division of Gastroenterology and Hepatology, Kansas City Veterans Affairs Medical Center, Kansas City, MO, USA
| | - Ken Robert Smith
- Department of Medicine (Gastroenterology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine (Genetic Epidemiology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Jathine Wong
- Department of Medicine (Gastroenterology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine (Genetic Epidemiology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Zhe Yu
- Department of Medicine (Gastroenterology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine (Genetic Epidemiology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Kenneth Boucher
- Department of Medicine (Gastroenterology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine (Genetic Epidemiology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Douglas O Faigel
- Division of Gastroenterology and Hepatology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Rahul Pannala
- Division of Gastroenterology and Hepatology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Randall W Burt
- Department of Medicine (Gastroenterology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine (Genetic Epidemiology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Karen Curtin
- Department of Medicine (Gastroenterology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Medicine (Genetic Epidemiology), Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - N Jewel Samadder
- Division of Gastroenterology and Hepatology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA.
| |
Collapse
|
48
|
Hidaka A, Sawada N, Svensson T, Goto A, Yamaji T, Shimazu T, Iwasaki M, Inoue M, Tsugane S. Family history of cancer and subsequent risk of cancer: A large‐scale population‐based prospective study in Japan. Int J Cancer 2019; 147:331-337. [DOI: 10.1002/ijc.32724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/11/2019] [Accepted: 09/20/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Akihisa Hidaka
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Norie Sawada
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Thomas Svensson
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
- Precision Health, Department of Bioengineering, Graduate School of EngineeringThe University of Tokyo Tokyo Japan
- Department of Clinical SciencesLund University, Skåne University Hospital Malmö Sweden
- Department of NeuropsychiatryKeio University School of Medicine Tokyo Japan
| | - Atsushi Goto
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Taiki Yamaji
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Taichi Shimazu
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Motoki Iwasaki
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Manami Inoue
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | - Shoichiro Tsugane
- Epidemiology and Prevention Group, Center for Public Health SciencesNational Cancer Center Tokyo Japan
| | | |
Collapse
|
49
|
Lo W, Morris MC, Ahmad SA, Patel SH. Screening patients at high risk for pancreatic cancer—Is it time for a paradigm shift? J Surg Oncol 2019; 120:851-857. [DOI: 10.1002/jso.25616] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/16/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Winifred Lo
- Department of SurgeryUniversity of Cincinnati College of MedicineCincinnati Ohio
| | - Mackenzie C. Morris
- Department of SurgeryUniversity of Cincinnati College of MedicineCincinnati Ohio
| | - Syed A. Ahmad
- Department of SurgeryUniversity of Cincinnati College of MedicineCincinnati Ohio
| | - Sameer H. Patel
- Department of SurgeryUniversity of Cincinnati College of MedicineCincinnati Ohio
| |
Collapse
|
50
|
Navarro EB, López EV, Quijano Y, Caruso R, Ferri V, Durand H, Cabrera IF, Reques ED, Ielpo B, Glagolieva AY, Plaza C. Impact of BRCA1/2 gene mutations on survival of patients with pancreatic cancer: A case-series analysis. Ann Hepatobiliary Pancreat Surg 2019; 23:200-205. [PMID: 31225426 PMCID: PMC6558134 DOI: 10.14701/ahbps.2019.23.2.200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/19/2022] Open
Abstract
BRCA gene mutations are found in up to 10% of pancreatic adenocarcinoma cases. We present a description of 4 cases along with a review of the current literature regarding pathogenesis, target treatment, response and survival rates in these types of malignancies. We describe four cases of pancreatic adenocarcinoma, in three of which the BRCA2 mutation was identified, in one - BRCA1 gene alteration. Two patients underwent surgery following the neoadjuvant treatment with Folfirinox and radiotherapy; in the first case, a distal pancreatectomy with splenectomy was performed and in the second one - the Whipple's procedure. In both cases, a complete pathological response was reported. Other 2 patients were treated with Folfirinox after BRCA mutation identification and acceptable life expectancy was obtained. The association of pathologic complete response (PCR) with lower rates of local recurrence and better survival in patients with various types of adenocarcinomas is well known. Identification of such patients carrying BRCA mutations could provide an application of better personalized treatment. In some patients with pancreatic cancer, especially when there is clinical or demographic reason to suspect a genetic predisposition, a confirmation of the presence of BRCA mutations could provide an opportunity to use target treatment with beneficial outcomes regarding survival.
Collapse
Affiliation(s)
- Ernesto Barzola Navarro
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Emilio Vicente López
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Yolanda Quijano
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Riccardo Caruso
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Valentina Ferri
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Hipolito Durand
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Isabel Fabra Cabrera
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Eduardo Diaz Reques
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | - Benedetto Ielpo
- Department of General and Digestive Surgery, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| | | | - Carlos Plaza
- Department of Pathology, Hospital Universitario Madrid Sanchinarro, Madrid, Spain
| |
Collapse
|