1
|
Askari N, Pirouz MS, Mafikandi V, Hadizadeh M, Mousavi SZ. The lncRNA UCA1 Enhances Pancreatic Cancer EMT by Regulating miR-708-5p and miR-135b-5p: A Bioinformatics Approach. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:1659-1669. [PMID: 39086404 PMCID: PMC11287582 DOI: 10.18502/ijph.v53i7.16060] [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: 08/24/2023] [Accepted: 11/11/2023] [Indexed: 08/02/2024]
Abstract
Background Pancreatic cancer (PC) is an exceedingly malignant ailment that is not only characterized by its insidious onset and rapid progression but also by its poor therapeutic effects. Recently, emerging evidence has shed light on the significant role that non-coding RNAs (ncRNAs), particularly long ncRNAs (lncRNAs) and microRNAs (miRNAs), play in the pathogenesis of PC. This investigation aimed to construct a network of interactions between miRNAs, lncRNAs, and mRNAs, as well as to perform correlation analyses in the context of PC. Methods This study carried out in Kerman City, southeastern Iran in 2023. We utilized the GSE119794 dataset from the Gene Expression Omnibus (GEO) to analyze differentially expressed lncRNAs (DE-lncRNAs), miRNAs (DE-miRNAs), and mRNAs (DE-mRNAs). Following the identification of the DE-lncRNAs, DE-mRNAs, and DE-miRNAs, we proceeded to examine differentially expressed epithelialmesenchymal transition (EMT) genes. Subsequently, we utilized the RNAInter database to predict interactions among lncRNAs, miRNAs, and mRNAs. Finally, we employed Cytoscape to visualize and analyze the constructed network. Results 14 DE-lncRNAs, 14 DE-miRNAs, 545 DE-mRNAs, and 65 DE-EMT from pancreatic cancer and its adjacent tissue RNA-Seq data were identified. 1184 EMT genes from dbEMT were obtained, among which 65 DE-EMT were assigned as EMT genes and correlated with tumor progression. One functional lncRNA (UCA1) was identified as a key functional lncRNA. The area under the ROC curve (AUC) of UCA1 and miR-708-5p were 0.79 and 0.86, respectively. Thus, it is reasonable to believe that this prognostic risk model helps predict PC metastasis. Conclusion UCA1 is a new lncRNA linked with EMT in PC and contributes to a better knowledge of the regulatory mechanisms related to lncRNAs in PC.
Collapse
Affiliation(s)
- Nahid Askari
- Department of Biotechnology, Institute of Sciences and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Marziye Shad Pirouz
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Vida Mafikandi
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyedeh Zahra Mousavi
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
2
|
Kim SY, Jo MJ, Yoon MS, Jin CE, Shin YB, Lee JM, Shin HJ, Oh JG, Cho JM, Kim H, Park H, Choi YW, Park CW, Kim JS, Shin DH. Gemcitabine and rapamycin-loaded mixed polymeric thermogel for metastatic pancreatic cancer therapy. J Control Release 2023; 360:796-809. [PMID: 37437850 DOI: 10.1016/j.jconrel.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/12/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer-related death and has a poor 5-year overall survival. The superior therapeutic benefits of combination or co-administration of drugs as intraperitoneal chemotherapy have increased interest in developing strategies to deliver chemotherapeutic agents to patients safely. In this study, we prepared a gel comprising the thermosensitive poly(lactide-co-glycolide)-b-poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PLGA-PEG-PLGA) polymer and gemcitabine (GEM), which is currently used as the primary chemotherapy for PDAC and rapamycin (RAPA), a mammalian TOR (mTOR) inhibitor, to deliver the drug through intraperitoneal injection. We performed in vitro cytotoxicity experiments to verify the synergistic effects of the two drugs at different molar ratios and characterized the physicochemical properties of the GEM, RAPA, and GEM/RAPA-loaded thermosensitive PLGA-PEG-PLGA gels, hereafter referred to as (g(G), g(R), and g(GR)), respectively. The g(GR) comprising PLGA-PEG-PLGA polymer (25% w/v) and GEM and RAPA at a molar ratio of 11:1 showed synergism and was optimized. An in vitro cytotoxicity assay was performed by treating Panc-1-luc2 tumor spheroids with g(G), g(R), or g(GR). The g(GR) treatment group showed a 2.75-fold higher inhibition rate than the non-treated (NT) and vehicle-treated groups. Furthermore, in vivo drug release assay in mice by intraperitoneal injection of g(G), g(R), or g(GR) showed a more rapid release rate of GEM than RAPA, similar to the in vitro release pattern. The drugs in the gel were released faster in vivo than in vitro and degraded in 48 h. In addition, g(GR) showed the highest anti-tumor efficacy with no toxicity to mice. These results provide evidence for the safety and efficacy of g(GR) for intraperitoneal drug delivery. This study will assist in developing and clinically administering topical anti-cancer formulations.
Collapse
Affiliation(s)
- Seo Yeon Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Min Jeong Jo
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Moon Sup Yoon
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Chae Eun Jin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yu Been Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jae Min Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Hee Ji Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Joon Gyo Oh
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Jae Min Cho
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Hyunjun Kim
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Hyunjin Park
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Yong-Won Choi
- R&D Center, Huons Co., Ltd., Ansan, 15588, Republic of Korea
| | - Chun-Woong Park
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jin-Seok Kim
- Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea.
| |
Collapse
|
3
|
Badheeb M, Abdelrahim A, Esmail A, Umoru G, Abboud K, Al-Najjar E, Rasheed G, Alkhulaifawi M, Abudayyeh A, Abdelrahim M. Pancreatic Tumorigenesis: Precursors, Genetic Risk Factors and Screening. Curr Oncol 2022; 29:8693-8719. [PMID: 36421339 PMCID: PMC9689647 DOI: 10.3390/curroncol29110686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer (PC) is a highly malignant and aggressive tumor. Despite medical advancement, the silent nature of PC results in only 20% of all cases considered resectable at the time of diagnosis. It is projected to become the second leading cause in 2030. Most pancreatic cancer cases are diagnosed in the advanced stages. Such cases are typically unresectable and are associated with a 5-year survival of less than 10%. Although there is no guideline consensus regarding recommendations for screening for pancreatic cancer, early detection has been associated with better outcomes. In addition to continued utilization of imaging and conventional tumor markers, clinicians should be aware of novel testing modalities that may be effective for early detection of pancreatic cancer in individuals with high-risk factors. The pathogenesis of PC is not well understood; however, various modifiable and non-modifiable factors have been implicated in pancreatic oncogenesis. PC detection in the earlier stages is associated with better outcomes; nevertheless, most oncological societies do not recommend universal screening as it may result in a high false-positive rate. Therefore, targeted screening for high-risk individuals represents a reasonable option. In this review, we aimed to summarize the pathogenesis, genetic risk factors, high-risk population, and screening modalities for PC.
Collapse
Affiliation(s)
- Mohamed Badheeb
- Internal Medicine Department, College of Medicine, Hadhramout University, Mukalla 50512, Yemen
| | | | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Correspondence: (A.E.); (M.A.)
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Karen Abboud
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ebtesam Al-Najjar
- Faculty of Medicine and Health Sciences, University of Science and Technology, Sana’a 15201, Yemen
| | - Ghaith Rasheed
- Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | | | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX 77030, USA
- Weill Cornell Medical College, New York, NY 14853, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Correspondence: (A.E.); (M.A.)
| |
Collapse
|
4
|
Iyengar D, Tatiparti K, Gavande NS, Sau S, Iyer AK. Nanomedicine for overcoming therapeutic and diagnostic challenges associated with pancreatic cancer. Drug Discov Today 2022; 27:1554-1559. [PMID: 35247592 DOI: 10.1016/j.drudis.2022.02.026] [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: 06/08/2021] [Revised: 02/14/2022] [Accepted: 02/28/2022] [Indexed: 11/29/2022]
Abstract
Pancreatic cancer is the second leading cause of cancer-related death in the USA. The 5-year survival rate for pancreatic cancer is as low as 10%, making it one of the most deadly cancers. This dismal prognosis is caused, in part, by the lack of early detection and screening options, leading to late-stage detection of the disease, at a point at which chemotherapy is no longer effective. However, nanoparticle (NP) drug delivery systems have increased the efficacy of chemotherapeutics by improving the targeting ability of drugs to the tumor site, while also decreasing the risk of local and systemic toxicity. Such efforts can contribute to the development of early diagnosis and routine screening tests, which will drastically improve the survival rates and prognosis of patients with pancreatic cancer.
Collapse
Affiliation(s)
- Disha Iyengar
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Katyayani Tatiparti
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA.
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
5
|
Dean J, Goldberg E, Michor F. Designing optimal allocations for cancer screening using queuing network models. PLoS Comput Biol 2022; 18:e1010179. [PMID: 35622852 PMCID: PMC9182689 DOI: 10.1371/journal.pcbi.1010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 06/09/2022] [Accepted: 05/07/2022] [Indexed: 11/19/2022] Open
Abstract
Cancer is one of the leading causes of death, but mortality can be reduced by detecting tumors earlier so that treatment is initiated at a less aggressive stage. The tradeoff between costs associated with screening and its benefit makes the decision of whom to screen and when a challenge. To enable comparisons across screening strategies for any cancer type, we demonstrate a mathematical modeling platform based on the theory of queuing networks designed for quantifying the benefits of screening strategies. Our methodology can be used to design optimal screening protocols and to estimate their benefits for specific patient populations. Our method is amenable to exact analysis, thus circumventing the need for simulations, and is capable of exactly quantifying outcomes given variability in the age of diagnosis, rate of progression, and screening sensitivity and intervention outcomes. We demonstrate the power of this methodology by applying it to data from the Surveillance, Epidemiology and End Results (SEER) program. Our approach estimates the benefits that various novel screening programs would confer to different patient populations, thus enabling us to formulate an optimal screening allocation and quantify its potential effects for any cancer type and intervention. We describe a mathematical modeling methodology that offers quantitative insights into the potential benefits of screening and other interventions on cancer mortality. Our queuing-theoretic approach represents a potentially useful alternative to more traditional modeling approaches, in that it can provide more detailed results and entirely circumvents the need for simulations. Our methodology can be widely applied to estimate costs and benefits of screening strategies. By providing a detailed example of our method applied to epidemiological data, we hope to encourage greater uptake of this methodology in the community.
Collapse
Affiliation(s)
- Justin Dean
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Evan Goldberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
- Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- The Ludwig Center at Harvard, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
6
|
Yu YN, Ding XY, Tian ZB, Zhao QX, Ding XL, Yin XY, Wang XW, Jing X, Liu H. Immunoglobulin G4-related disease concomitant with pancreatic cancer: A rare case report. J Dig Dis 2022; 23:345-348. [PMID: 35751622 DOI: 10.1111/1751-2980.13106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 12/11/2022]
Affiliation(s)
- Ya Nan Yu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiao Yan Ding
- School of Basic Medicine, Qingdao University, Qingdao, Shandong Province, China
- Institute of Stem Cell and Regenerative Medicine, Qingdao University, Qingdao, Shandong Province, China
| | - Zi Bin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Qing Xi Zhao
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xue Li Ding
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiao Yan Yin
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xiao Wei Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Xue Jing
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | - Hua Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
7
|
Xu X, Song R, Hu K, Li Y, Jin H, Chen B, Song W, Zhang Y, Xu J, Sun Y. Multidisciplinary management for Peutz-Jeghers syndrome and prevention of vertical transmission to offspring using preimplantation genetic testing. Orphanet J Rare Dis 2022; 17:64. [PMID: 35189935 PMCID: PMC8862355 DOI: 10.1186/s13023-022-02221-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 02/06/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Peutz Jeghers syndrome (PJS) is an autosomal dominant genetic disorder caused by STK11 mutation with a predisposition to gastrointestinal polyposis and cancer. PJS patients suffer poor quality of life and are highly concerned about whether deleterious mutations transmit to their offspring. Therefore, this study aimed to propose feasible clinical management and provide effective preimplantation genetic testing for monogenic defect (PGT-M) strategies to protect offspring from inheriting the disease. METHODS A hospital-based clinical retrospective analysis reviewing the clinical characteristics and fertility aspects was first conducted on 51 PJS patients at the First Affiliated Hospital of Zhengzhou University between January 2016 and March 2021. Among the 51 patients, the PGT-M strategy was further carried out in 4 couples, which started with a biopsy of the trophectoderm cells of embryos and whole genome amplification using multiple displacement amplification. Thereafter, single nucleotide polymorphism linkage analyses based on karyomapping were performed with copy number variations of the embryos identified simultaneously. Finally, prenatal diagnosis was used to verify the validity of the PGT-M results. RESULTS A comprehensive management flowchart adopted by the multidisciplinary team model was formulated mainly focusing on clinical genetic and gastrointestinal aspects. Under the guidelines of this management, 32 embryos from 4 PJS pedigrees were diagnosed and 2 couples successfully conceived healthy babies free of the STK11 pathogenic mutation. CONCLUSIONS Our comprehensive management could help affected families avoid having children with PJS through preimplantation genetic testing and provide meaningful guidance for multidisciplinary clinical practice on PJS.
Collapse
Affiliation(s)
- Xiqiao Xu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruifeng Song
- Department of Gastroenterology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaiyue Hu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya Li
- Department of Gastroenterology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haixia Jin
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bing Chen
- Department of Gastroenterology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenyan Song
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yile Zhang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiawei Xu
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yingpu Sun
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Henan Engineering Laboratory of Preimplantation Genetic Diagnosis and Screening, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
8
|
Yang Y, Su X, Shen K, Zhang C, Dai H, Ma H, Jiang Y, Shuai L, Liu Z, You J, Min K, Chen Z. PUM1 is upregulated by DNA methylation to suppress antitumor immunity and results in poor prognosis in pancreatic cancer. Transl Cancer Res 2022; 10:2153-2168. [PMID: 35116535 PMCID: PMC8798770 DOI: 10.21037/tcr-20-3295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/26/2021] [Indexed: 12/24/2022]
Abstract
Background Pancreatic carcinoma (PAAD) is a highly malignant cancer with a poor prognosis and high mortality rate. Pumilio homologous protein 1 (PUM1) promotes cell growth, invasion, and metastasis and suppresses apoptosis in many different kinds of cancers, such as non-small-cell lung carcinoma (NSCLC), ovarian cancer and lymphocyte leukemia. However, the underlying mechanism and potential role of PUM1 in PAAD have not been investigated. Methods Bioinformatics analysis was performed using multiple databases [The Cancer Genome Atlas (TCGA), Gene Expression Profiling Interactive Analysis (GEPIA), BBCancer, Human Protein Atlas (HPA), MethSurv, cBioPortal, The Cancer Imaging Archive (TCIA), xCell, Gene Expression Omnibus (GEO)] to explore the diagnostic and prognostic role of PUM1, and the relationship between expression of PUM1 and prognosis of patients with PAAD. The analysis was further validated using the Kaplan-Meier plotter. Results PUM1 plays a role in both diagnostic and prognostic prediction. The PUM1 mRNA expression level correlates with both the prognosis and incidence of pancreatic cancer. PUM1 can serve as a potential diagnostic indicator for pancreatic cancer. Furthermore, the DNA methylation levels of PUM1 affects its oncogene function in pancreatic cancer. PUM1 can also inhibit the immune microenvironment by altering immune cell infiltration, which affects immunotherapy response in pancreatic cancer. Conclusions PUM1 takes a crucial part in the immune microenvironment and immunotherapy response of PAAD and is potentially useful for the development of novel diagnostic and treatment strategies.
Collapse
Affiliation(s)
- Yishi Yang
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Xingxing Su
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Kaicheng Shen
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Chengcheng Zhang
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Haisu Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Hongbo Ma
- Department of Oncology, The Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Yan Jiang
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Ling Shuai
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Zhipeng Liu
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Jinshan You
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Ke Min
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Zhiyu Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, First Affiliated Hospital of the Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Gheorghe G, Diaconu CC, Ionescu V, Constantinescu G, Bacalbasa N, Bungau S, Gaman MA, Stan-Ilie M. Risk Factors for Pancreatic Cancer: Emerging Role of Viral Hepatitis. J Pers Med 2022; 12:83. [PMID: 35055398 PMCID: PMC8780367 DOI: 10.3390/jpm12010083] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/09/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most aggressive malignant neoplastic diseases. The incidence and mortality rates of this disease vary depending on geographical area, which might be explained by the different exposure to risk factors. To improve the prognosis of patients with pancreatic cancer, different approaches are needed for an earlier diagnosis. Identification of risk factors and implementation of screening strategies are essential for a better prognosis. Currently, the risk factors for pancreatic cancer fall into two broad categories, namely extrinsic and intrinsic factors. Extrinsic factors include alcohol consumption, smoking, a diet rich in saturated fats, and viral infections such as chronic infection with hepatitis B and C viruses. The pathophysiological mechanisms explaining how these hepatotropic viruses contribute to the development of pancreatic cancer are not fully elucidated. The common origin of hepatocytes and pancreatic cells in the multipotent endodermal cells, the common origin of the blood vessels and biliary ducts of the pancreas and the liver, or chronic inflammatory changes may be involved in this interaction. A careful monitoring of patients with viral liver infections may contribute to the early diagnosis of pancreatic cancer and improve the prognosis of these patients.
Collapse
Affiliation(s)
- Gina Gheorghe
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| | - Camelia Cristina Diaconu
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Vlad Ionescu
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| | - Gabriel Constantinescu
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| | - Nicolae Bacalbasa
- Department of Visceral Surgery, “Carol Davila” University of Medicine and Pharmacy, Center of Excellence in Translational Medicine “Fundeni” Clinical Institute, 022328 Bucharest, Romania;
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - Mihnea-Alexandru Gaman
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Madalina Stan-Ilie
- Department 5, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (G.C.); (M.-A.G.); (M.S.-I.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania;
| |
Collapse
|
10
|
Chen Y, Liu Y, Tao Q, Fan Y, Ma C, Li D, Huang F, Tang D. circPUM1 Activates the PI3K/AKT Signaling Pathway by Sponging to Promote the Proliferation, Invasion and Glycolysis of Pancreatic Cancer. Curr Pharm Biotechnol 2022; 23:1405-1414. [PMID: 34259142 DOI: 10.2174/1389201022666210713152457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/28/2021] [Accepted: 06/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Our study seeks to obtain data to assess the impact of circPUM1 on pancreatic cancer (PC) and its mechanism. METHODS The expression of circPUM1 and miR-200c-3p in PC and normal tissues and PC cell lines was collected and detected, and subsequently dual-luciferase assay-based verification of the binding site of the two was carried out. After interfering with circPUM1 expression in MIAPaCa-2 and PANC-1 cells, cell proliferation, viability, apoptosis rate, invasion ability, glucose consumption, and lactate production were measured by MTT, colony formation, flow cytometry, Transwell assays, and glucose and lactate assay kits. Additionally, western blot was utilized for assessing PI3K/AKT signaling pathway-related proteins. From the results, highly expressed circPUM1 and miR-200c-3p in PC tissues and cells were proved. RESULTS Down-regulation of circPUM1 expression significantly inhibited cell proliferation, cell viability, invasion and glycolysis, while increasing the apoptosis rate. Down-regulated circPUM1 led to the inhibition of the PI3K/AKT signaling pathway activity in PC cells; while up-regulated circPUM1 increased its activity. Further experiments revealed that down-regulation of miR-200c-3p expression reversed the inhibitory effect of lowly expressed circPUM1 on PC cells. CONCLUSION In summary, circPUM1 activates PI3K/AKT signaling pathway by sponging miR-200c-3p and promotes PC progression.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gastrointestinal Hepatobiliary Surgery, Shenzhen Longhua District Central Hospital, ShenZhen, Guangdong 510075, China
| | - Yuhong Liu
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| | - Qiang Tao
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| | - YouWen Fan
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| | - Chao Ma
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| | - Dong Li
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| | - Fei Huang
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| | - Di Tang
- Department of General Surgery, The Seventh Affiliated Hospital, Sen Yat-sen University, ShenZhen, Guangdong, 518017, China
| |
Collapse
|
11
|
Lew D, Kwok K. Diagnosis and Evaluation of Pancreatic and Periampullary Adenocarcinoma. HEPATO-PANCREATO-BILIARY MALIGNANCIES 2022:431-459. [DOI: 10.1007/978-3-030-41683-6_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Casà C, Piras A, D’Aviero A, Preziosi F, Mariani S, Cusumano D, Romano A, Boskoski I, Lenkowicz J, Dinapoli N, Cellini F, Gambacorta MA, Valentini V, Mattiucci GC, Boldrini L. The impact of radiomics in diagnosis and staging of pancreatic cancer. Ther Adv Gastrointest Endosc 2022; 15:26317745221081596. [PMID: 35342883 PMCID: PMC8943316 DOI: 10.1177/26317745221081596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 02/02/2022] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Pancreatic cancer (PC) is one of the most aggressive tumours, and better risk stratification among patients is required to provide tailored treatment. The meaning of radiomics and texture analysis as predictive techniques are not already systematically assessed. The aim of this study is to assess the role of radiomics in PC. METHODS A PubMed/MEDLINE and Embase systematic review was conducted to assess the role of radiomics in PC. The search strategy was 'radiomics [All Fields] AND ("pancreas" [MeSH Terms] OR "pancreas" [All Fields] OR "pancreatic" [All Fields])' and only original articles referred to PC in humans in the English language were considered. RESULTS A total of 123 studies and 183 studies were obtained using the mentioned search strategy on PubMed and Embase, respectively. After the complete selection process, a total of 56 papers were considered eligible for the analysis of the results. Radiomics methods were applied in PC for assessment technical feasibility and reproducibility aspects analysis, risk stratification, biologic or genomic status prediction and treatment response prediction. DISCUSSION Radiomics seems to be a promising approach to evaluate PC from diagnosis to treatment response prediction. Further and larger studies are required to confirm the role and allowed to include radiomics parameter in a comprehensive decision support system.
Collapse
Affiliation(s)
- Calogero Casà
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Andrea D’Aviero
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Preziosi
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Mariani
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Davide Cusumano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Angela Romano
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ivo Boskoski
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCSS, Rome, Italy
| | - Jacopo Lenkowicz
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Nicola Dinapoli
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Cellini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Antonietta Gambacorta
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Carlo Mattiucci
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Boldrini
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
13
|
Spiliopoulos S, Zurlo MT, Casella A, Laera L, Surico G, Surgo A, Fiorentino A, de'Angelis N, Calbi R, Memeo R, Inchingolo R. Current status of non-surgical treatment of locally advanced pancreatic cancer. World J Gastrointest Oncol 2021; 13:2064-2075. [PMID: 35070042 PMCID: PMC8713317 DOI: 10.4251/wjgo.v13.i12.2064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/28/2021] [Accepted: 10/25/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the 7th leading cause of death due to cancer in industrialized countries and the 11th most common cancer globally, with 458918 new cases (2.5% of all cancers) and 432242 deaths (4.5% of all cancer deaths) in 2018. Unfortunately, 80% to 90% of the patients present with unresectable disease, and the reported 5-year survival rate range between 10% and 25%, even after successful resection with tumor-free margins. Systemic chemotherapy, radiotherapy, and minimally invasive image-guided procedures that have emerged over the past years, are used for the management of non-operable PC. This review focuses on currently available non-surgical options of locally advanced pancreatic cancer.
Collapse
Affiliation(s)
- Stavros Spiliopoulos
- 2nd Radiology Department, Interventional Radiology Unit, National and Kapodistrian University of Athens, Athens 12461, Greece
| | - Maria Teresa Zurlo
- Interventional Radiology Unit, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| | - Annachiara Casella
- Unit of Hepato-Pancreatic-Biliary Surgery, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| | - Letizia Laera
- Department of Oncology, General Regional Hospital “F. Miulli”, Acquaviva delle Fonti 70021, Italy
| | - Giammarco Surico
- Department of Oncology, General Regional Hospital “F. Miulli”, Acquaviva delle Fonti 70021, Italy
| | - Alessia Surgo
- Department of Radiation Oncology, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| | - Alba Fiorentino
- Department of Radiation Oncology, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| | - Nicola de'Angelis
- Unit of Minimally Invasive and Robotic Digestive Surgery, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| | - Roberto Calbi
- Department of Radiology, General Regional Hospital “F. Miulli”, Acquaviva delle Fonti 70021, Italy
| | - Riccardo Memeo
- Unit of Hepato-Pancreatic-Biliary Surgery, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| | - Riccardo Inchingolo
- Interventional Radiology Unit, “F. Miulli” Regional General Hospital, Acquaviva delle Fonti 70021, Italy
| |
Collapse
|
14
|
Park MY, Lee W, Kwon J, Song KB, Hwang DW, Lee JH, Kim SC. Comparison of perioperative outcomes in pancreatic head cancer patients following either a laparoscopic or open pancreaticoduodenectomy with a superior mesenteric artery first approach. Ann Hepatobiliary Pancreat Surg 2021; 25:358-365. [PMID: 34402436 PMCID: PMC8382868 DOI: 10.14701/ahbps.2021.25.3.358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 02/24/2021] [Accepted: 03/02/2021] [Indexed: 12/30/2022] Open
Abstract
Backgrounds/Aims A superior mesenteric artery first approach (SFA) technique can improve the complete resection rate. It can be used to determine whether an operation can be performed by invading the superior mesenteric artery before performing a pancreatic transection in patients with pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to compare perioperative outcomes between laparoscopic and open SFA for PDAC. Methods Between January 2017 and August 2019, consecutive patients who underwent laparoscopic and open pancreaticoduodenectomy (PD) for PDAC using SFA procedures were included and compared between laparoscopic and open procedures. Results Fourteen and 83 patients underwent laparoscopic and open surgeries, respectively. In perioperative outcomes, there were no significant differences in the amount of intraoperative blood loss or transfusion rate between the two groups. In the laparoscopic group, the operation time was longer with less patients showing wound infection. R0 resection rate and the number of retrieved lymph nodes showed no significant difference. The average time to adjuvant chemotherapy was longer in the open group. There was no significant difference in the mean survival time or the recurrence free period. Conclusions Patients who underwent laparoscopic PD using SFA showed perioperative outcomes comparable compared to those of patients who underwent open procedures performed by experienced surgeons.
Collapse
Affiliation(s)
- Min Young Park
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woohyung Lee
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaewoo Kwon
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ki Byung Song
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dae Wook Hwang
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hoon Lee
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Song Cheol Kim
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Gupta N, Yelamanchi R. Pancreatic adenocarcinoma: A review of recent paradigms and advances in epidemiology, clinical diagnosis and management. World J Gastroenterol 2021; 27:3158-3181. [PMID: 34163104 PMCID: PMC8218366 DOI: 10.3748/wjg.v27.i23.3158] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/03/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the dreaded malignancies for both the patient and the clinician. The five-year survival rate of pancreatic adenocarcinoma (PDA) is as low as 2% despite multimodality treatment even in the best hands. As per the Global Cancer Observatory of the International Agency for Research in Cancer estimates of pancreatic cancer, by 2040, a 61.7% increase is expected in the total number of cases globally. With the widespread availability of next-generation sequencing, the entire genome of the tumors is being sequenced regularly, providing insight into their pathogenesis. As invasive PDA arises from pancreatic intraepithelial neoplasia and mucinous neoplasm and intraductal papillary neoplasm, screening for them can be beneficial as the disease is curable with resection at an early stage. Routine preoperative biliary drainage has no role in patients suffering from PDA with obstructive jaundice. If performed, metallic stents are preferred over plastic ones. Minimally invasive procedures are preferred to open procedures as they have less morbidity. The duct-to-mucosa technique for pancreaticojejunostomy is presently widely practiced. The role of intraperitoneal drains after surgery for PDA is controversial. Neoadjuvant chemoradiotherapy has been proven to have a significant role both in locally advanced as well as in resectable PDA. Many new regimens and drugs have been added in the arsenal of chemoradiotherapy for metastatic disease. The roles of immunotherapy and gene therapy in PDA are being investigated. This review article is intended to improve the understanding of the readers with respect to the latest updates of PDA, which may help to trigger new research ideas and make better management decisions.
Collapse
Affiliation(s)
- Nikhil Gupta
- Department of Surgery, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, Delhi 110001, India
| | - Raghav Yelamanchi
- Department of Surgery, Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, Delhi 110001, India
| |
Collapse
|
16
|
Gheorghe G, Bungau S, Ilie M, Behl T, Vesa CM, Brisc C, Bacalbasa N, Turi V, Costache RS, Diaconu CC. Early Diagnosis of Pancreatic Cancer: The Key for Survival. Diagnostics (Basel) 2020; 10:869. [PMID: 33114412 PMCID: PMC7694042 DOI: 10.3390/diagnostics10110869] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most aggressive forms of cancer. Negative prognosis is mainly due to the late diagnosis in advanced stages, when the disease is already therapeutically overcome. Studies in recent years have focused on identifying biomarkers that could play a role in early diagnosis, leading to the improvement of morbidity and mortality. Currently, the only biomarker widely used in the diagnosis of PC is carbohydrate antigen 19-9 (CA19.9), which has, however, more of a prognostic role in the follow-up of postoperative recurrence than a diagnostic role. Other biomarkers, recently identified as the methylation status of ADAMTS1 (A disintegrin and metalloproteinase with thrombospondin motifs 1) and BNC1 (zinc finger protein basonuclin-1) in cell-free deoxyribonucleic acid (DNA), may play a role in the early detection of PC. This review focuses on the diagnosis of PC in its early stages.
Collapse
Affiliation(s)
- Gina Gheorghe
- Department 5, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (M.I.); (R.S.C.)
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Madalina Ilie
- Department 5, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (M.I.); (R.S.C.)
- Department of Gastroenterology, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Cosmin Mihai Vesa
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410041 Oradea, Romania;
| | - Ciprian Brisc
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410041 Oradea, Romania;
| | - Nicolae Bacalbasa
- Department of Surgery, “Ion Cantacuzino” Clinical Hospital, 030167 Bucharest, Romania;
- Department 13, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Vladiana Turi
- Department of Cardiology, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Raluca Simona Costache
- Department 5, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (M.I.); (R.S.C.)
- Department of Gastroenterology, “Carol Davila” University Emergency Central Military Hospital, 010825 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Department 5, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (G.G.); (M.I.); (R.S.C.)
- Department of Internal Medicine, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| |
Collapse
|
17
|
Jin Z, Chen Q, Tan L, Jia B, Liu Y. WITHDRAWN: Novel long-acting BF-30 conjugate corrects pancreatic carcinoma via cytoplasmic membrane permeabilization and DNA-binding in tumor-bearing mice. Life Sci 2020:118278. [PMID: 32798555 DOI: 10.1016/j.lfs.2020.118278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
This article has been withdrawn at the request of the editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Zhe Jin
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Qingmin Chen
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ludong Tan
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Baoxing Jia
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yahui Liu
- The First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
18
|
Jiang N, Dai Q, Su X, Fu J, Feng X, Peng J. Role of PI3K/AKT pathway in cancer: the framework of malignant behavior. Mol Biol Rep 2020; 47:4587-4629. [PMID: 32333246 PMCID: PMC7295848 DOI: 10.1007/s11033-020-05435-1] [Citation(s) in RCA: 367] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/03/2020] [Indexed: 12/12/2022]
Abstract
Given that the PI3K/AKT pathway has manifested its compelling influence on multiple cellular process, we further review the roles of hyperactivation of PI3K/AKT pathway in various human cancers. We state the abnormalities of PI3K/AKT pathway in different cancers, which are closely related with tumorigenesis, proliferation, growth, apoptosis, invasion, metastasis, epithelial-mesenchymal transition, stem-like phenotype, immune microenvironment and drug resistance of cancer cells. In addition, we investigated the current clinical trials of inhibitors against PI3K/AKT pathway in cancers and found that the clinical efficacy of these inhibitors as monotherapy has so far been limited despite of the promising preclinical activity, which means combinations of targeted therapy may achieve better efficacies in cancers. In short, we hope to feature PI3K/AKT pathway in cancers to the clinic and bring the new promising to patients for targeted therapies.
Collapse
Affiliation(s)
- Ningni Jiang
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Qijie Dai
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Xiaorui Su
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Jianjiang Fu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Xuancheng Feng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
| | - Juan Peng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Guangzhou, 510150 China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, 510150 China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, 510150 China
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
| |
Collapse
|
19
|
Ethnic and racial disparities of pancreatic adenocarcinoma in Florida. HPB (Oxford) 2020; 22:735-743. [PMID: 31601507 DOI: 10.1016/j.hpb.2019.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/18/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Racial disparities are known to negatively impact survival in patients with pancreatic adenocarcinoma. However, data regarding the Hispanic ethnicity are scarce in the pancreatic cancer literature. Therefore, the aim of this study is to analyze whether race and ethnicity are independent predictors of survival in patients with pancreatic adenocarcinoma in Florida. METHODS A retrospective study was performed utilizing all patients diagnosed with pancreatic adenocarcinoma between 1983 and 2013 in the Florida Cancer Data System (FCDS). Statistical analysis was performed using Cox proportional hazard regression models, and Kaplan-Meier survival analysis. RESULTS Of 36,756 patients identified with pancreatic adenocarcinoma in the FCDS, 9.1% were Hispanic and 91% were non-Hispanic. Ethnicity was associated with improved survival among Hispanics compared to non-Hispanics (HR 0.86, 95% CI 0.82-0.90, both p = 0.001). Furthermore, 90% of patients were White, and 9% were Black. Compared to Whites, Blacks had a significantly decreased survival (HR 1.07, 95% CI 1.03-1.13, p = 0.003). CONCLUSION In Florida patients with pancreatic adenocarcinoma, Hispanic ethnicity is associated with improved survival compared to Non-Hispanics. Additionally, Blacks present at an earlier age and later stage of diagnosis with worse survival compared to Whites and Others.
Collapse
|
20
|
Koo DH, Han KD, Kim HJ, Park CY. Middle-aged men with type 2 diabetes as potential candidates for pancreatic cancer screening: a 10-year nationwide population-based cohort study. Acta Diabetol 2020; 57:197-202. [PMID: 31420738 DOI: 10.1007/s00592-019-01405-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/08/2019] [Indexed: 01/08/2023]
Abstract
AIMS Before developing a national screening program for pancreatic cancer, more detailed and reliable estimation of pancreatic cancer incidence rate is needed according to sex, age, and diabetes mellitus status. METHODS Among populations who underwent a biennial or annual evaluation provided by the National Health Insurance Service Database of Claims between 2006 and 2015, data were evaluated from the medical records of 34.2 million individuals aged 30 years and over. RESULTS The annual incidence rate (IR; per 100,000) of pancreatic cancer in 2006 was 5.96, and the IR in 2015 increased to 8.92. The IRs increased consistently and significantly with age (p for trend < 0.0001). The incidence rate ratio (IRR; after adjusting for age and sex) of pancreatic cancer was higher (about 1.5 times) in males than in females for all ages. In particular, the IR (15.8) of pancreatic cancer between 50 and 59 years in patients with diabetes was strikingly higher compared to that in patients without diabetes (IR 7.6). Furthermore, the pancreatic cancer IR (19.1) of men with diabetes between 50 and 59 years was similar to the IR (20.2) in women with diabetes between 60 and 69 years, and it was even greater than the IR (17.0) in non-diabetic women between 60 and 69 years. CONCLUSIONS In middle-aged populations, men with type 2 diabetes had the highest relative risk. Their annual incidence rate of pancreatic cancer persistently increased and was higher by about 10-30% than in women with diabetes.
Collapse
Affiliation(s)
- Dong-Hoe Koo
- Division of Hematology/Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung-Do Han
- Department of Biostatistics, Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hong Joo Kim
- Division of Gastroenterology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Cheol-Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, 29 Saemunan-ro, Jongno-gu, Seoul, 03181, Korea.
| |
Collapse
|
21
|
Chen Q, Yang C, Chen L, Zhang JJ, Ge WL, Yuan H, Meng LD, Huang XM, Shen P, Miao Y, Jiang KR. YY1 targets tubulin polymerisation-promoting protein to inhibit migration, invasion and angiogenesis in pancreatic cancer via p38/MAPK and PI3K/AKT pathways. Br J Cancer 2019; 121:912-921. [PMID: 31631174 PMCID: PMC6888832 DOI: 10.1038/s41416-019-0604-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/21/2019] [Accepted: 09/27/2019] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PDAC) is a highly invasive cancer with poor prognosis. Recent research has found that the transcription factor Yin Yang 1 (YY1) plays an inhibitory role in the development of pancreatic cancer. It has been reported that tubulin polymerisation-promoting protein (TPPP) plays an indispensable role in a variety of tumours, but its expression and role in pancreatic cancer have not yet been elucidated. METHODS In this study, we performed ChIP-sequencing and found that YY1 directly binds to the promoter region of TPPP. The expression of TPPP in pancreatic cancer was detected by western blotting and immunohistochemistry. Four-week-old male BALB/c-nude mice were used to assess the effect of TPPP on pancreatic cancer. RESULTS Immunohistochemistry revealed that TPPP was expressed at low levels in pancreatic cancer tissues, and was associated with blood vessel invasion. The results from vivo experiments have showed that TPPP could enhance the migration and invasion of pancreatic cancer. Further experiments showed that YY1 could inhibit the migration, invasion and angiogenesis of pancreatic cancer cells by downregulating TPPP via p38/MAPK and PI3K/AKT pathways. CONCLUSION Our study demonstrates that TPPP may act as a promoter and may serve as a novel target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Qun Chen
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Chuang Yang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Lei Chen
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Jing-Jing Zhang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Wan-Li Ge
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Hao Yuan
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Ling-Dong Meng
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Xu-Min Huang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Peng Shen
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yi Miao
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Pancreas Institute, Nanjing Medical University, Nanjing, China.
| | - Kui-Rong Jiang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Pancreas Institute, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
22
|
Dalal V, Carmicheal J, Dhaliwal A, Jain M, Kaur S, Batra SK. Radiomics in stratification of pancreatic cystic lesions: Machine learning in action. Cancer Lett 2019; 469:228-237. [PMID: 31629933 DOI: 10.1016/j.canlet.2019.10.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/03/2019] [Accepted: 10/15/2019] [Indexed: 12/15/2022]
Abstract
Pancreatic cystic lesions (PCLs) are well-known precursors of pancreatic cancer. Their diagnosis can be challenging as their behavior varies from benign to malignant disease. Precise and timely management of malignant pancreatic cysts might prevent transformation to pancreatic cancer. However, the current consensus guidelines, which rely on standard imaging features to predict cyst malignancy potential, are conflicting and unclear. This has led to an increased interest in radiomics, a high-throughput extraction of comprehensible data from standard of care images. Radiomics can be used as a diagnostic and prognostic tool in personalized medicine. It utilizes quantitative image analysis to extract features in conjunction with machine learning and artificial intelligence (AI) methods like support vector machines, random forest, and convolutional neural network for feature selection and classification. Selected features can then serve as imaging biomarkers to predict high-risk PCLs. Radiomics studies conducted heretofore on PCLs have shown promising results. This cost-effective approach would help us to differentiate benign PCLs from malignant ones and potentially guide clinical decision-making leading to better utilization of healthcare resources. In this review, we discuss the process of radiomics, its myriad applications such as diagnosis, prognosis, and prediction of therapy response. We also discuss the outcomes of studies involving radiomic analysis of PCLs and pancreatic cancer, and challenges associated with this novel field along with possible solutions. Although these studies highlight the potential benefit of radiomics in the prevention and optimal treatment of pancreatic cancer, further studies are warranted before incorporating radiomics into the clinical decision support system.
Collapse
Affiliation(s)
- Vipin Dalal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Joseph Carmicheal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amaninder Dhaliwal
- Department of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; The Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; The Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
23
|
Christodoulou E, Visser M, Potjer TP, van der Stoep N, Rodríguez-Girondo M, van Doorn R, Gruis N. Assessing a single SNP located at TERT/CLPTM1L multi-cancer risk region as a genetic modifier for risk of pancreatic cancer and melanoma in Dutch CDKN2A mutation carriers. Fam Cancer 2019; 18:439-444. [PMID: 31203567 PMCID: PMC6784815 DOI: 10.1007/s10689-019-00137-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Carriers of pathogenic variants in CDKN2A have a 70% life-time risk of developing melanoma and 15–20% risk of developing pancreatic cancer (PC). In the Netherlands, a 19-bp deletion in exon 2 of CDKN2A (p16-Leiden mutation) accounts for most hereditary melanoma cases. Clinical experience suggests variability in occurrence of melanoma and PC in p16-Leiden families. Thereby, the risk of developing cancer could be modified by both environmental and genetic contributors, suggesting that identification of genetic modifiers could improve patients’ surveillance. In a recent genome-wide association study (GWAS), rs36115365-C was found to significantly modify risk of PC and melanoma in the European population. This SNP is located on chr5p15.33 and has allele-specific regulatory activities on TERT expression. Herein, we investigated the modifying capacities of rs36115365-C on PC and melanoma in a cohort of 283 p16-Leiden carriers including 29 diagnosed with PC, 171 diagnosed with melanoma, 21 diagnosed with both PC and melanoma and 62 with neither PC nor melanoma. In contrast to previously reported findings, we did not find a significant association of PC risk with risk variant presence as determined by Generalized Estimating Equations (GEE) modelling. Interestingly, carrier-ship of the risk variant had a significant protective effect for melanoma (OR − 0.703 [95% CI − 1.201 to − 0.205], p = 0.006); however, the observed association was no longer significant after exclusion of probands to assess possible influence of ascertainment. Collectively, genetic modifiers for the prediction of PC and melanoma risk in p16-Leiden carriers remain to be determined.
Collapse
Affiliation(s)
- E Christodoulou
- Department of Dermatology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - M Visser
- Department of Dermatology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - T P Potjer
- Department of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - N van der Stoep
- Department of Clinical Genetics, LUMC, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - M Rodríguez-Girondo
- Section of Medical Statistics, Department of Biomedical Data Sciences, LUMC, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - R van Doorn
- Department of Dermatology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - N Gruis
- Department of Dermatology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
24
|
Eissa MAL, Lerner L, Abdelfatah E, Shankar N, Canner JK, Hasan NM, Yaghoobi V, Huang B, Kerner Z, Takaesu F, Wolfgang C, Kwak R, Ruiz M, Tam M, Pisanic TR, Iacobuzio-Donahue CA, Hruban RH, He J, Wang TH, Wood LD, Sharma A, Ahuja N. Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood. Clin Epigenetics 2019; 11:59. [PMID: 30953539 PMCID: PMC6451253 DOI: 10.1186/s13148-019-0650-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 03/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite improvements in cancer management, most pancreatic cancers are still diagnosed at an advanced stage. We have recently identified promoter DNA methylation of the genes ADAMTS1 and BNC1 as potential blood biomarkers of pancreas cancer. In this study, we validate this biomarker panel in peripheral cell-free tumor DNA of patients with pancreatic cancer. RESULTS Sensitivity and specificity for each gene are as follows: ADAMTS1 87.2% and 95.8% (AUC = 0.91; 95% CI 0.71-0.86) and BNC1 64.1% and 93.7% (AUC = 0.79; 95% CI 0.63-0.78). When using methylation of either gene as a combination panel, sensitivity increases to 97.3% and specificity to 91.6% (AUC = 0.95; 95% CI 0.77-0.90). Adding pre-operative CA 19-9 values to the combined two-gene methylation panel did not improve sensitivity. Methylation of ADAMTS1 was found to be positive in 87.5% (7/8) of stage I, 77.8% (7/9) of stage IIA, and 90% (18/20) of stage IIB disease. Similarly, BNC1 was positive in 62.5% (5/8) of stage I patients, 55.6% (5/9) of stage IIA, and 65% (13/20) of patients with stage IIB disease. The two-gene panel (ADAMTS1 and/or BNC1) was positive in 100% (8/8) of stage I, 88.9% (8/9) of stage IIA, and 100% (20/20) of stage IIB disease. The sensitivity and specificity of the two-gene panel for localized pancreatic cancer (stages I and II), where the cancer is eligible for surgical resection with curative potential, was 94.8% and 91.6% respectively. Additionally, the two-gene panel exhibited an AUC of 0.95 (95% CI 0.90-0.98) compared to 57.1% for CA 19-9 alone. CONCLUSION The methylation status of ADAMTS1 and BNC1 in cfDNA shows promise for detecting pancreatic cancer during the early stages when curative resection of the tumor is still possible. This minimally invasive blood-based biomarker panel could be used as a promising tool for diagnosis and screening in a select subset of high-risk populations.
Collapse
Affiliation(s)
- Maryam A L Eissa
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lane Lerner
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eihab Abdelfatah
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nakul Shankar
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joseph K Canner
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nesrin M Hasan
- Department of Surgery, Yale-New Haven Health, Yale University, School of Medicine, P.O. Box 208062, New Haven, CT, 06520-8062, USA
| | - Vesal Yaghoobi
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barry Huang
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary Kerner
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Felipe Takaesu
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Wolfgang
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruby Kwak
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Ruiz
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Tam
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas R Pisanic
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA
| | - Christine A Iacobuzio-Donahue
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA.,Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Sol Goldman Pancreatic Cancer Research Center, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Jin He
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tza-Huei Wang
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, USA
| | - Laura D Wood
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,The Sol Goldman Pancreatic Cancer Research Center, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Anup Sharma
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Surgery, Yale-New Haven Health, Yale University, School of Medicine, P.O. Box 208062, New Haven, CT, 06520-8062, USA
| | - Nita Ahuja
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,The Sol Goldman Pancreatic Cancer Research Center, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA. .,Department of Surgery, Yale-New Haven Health, Yale University, School of Medicine, P.O. Box 208062, New Haven, CT, 06520-8062, USA.
| |
Collapse
|
25
|
Søreide K, Primavesi F, Labori KJ, Watson MM, Stättner S. Molecular biology in pancreatic ductal adenocarcinoma: implications for future diagnostics and therapy. Eur Surg 2019. [DOI: 10.1007/s10353-019-0575-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Rawla P, Sunkara T, Gaduputi V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J Oncol 2019; 10:10-27. [PMID: 30834048 PMCID: PMC6396775 DOI: 10.14740/wjon1166] [Citation(s) in RCA: 1462] [Impact Index Per Article: 243.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/14/2018] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide. However, its toll is higher in more developed countries. Reasons for vast differences in mortality rates of pancreatic cancer are not completely clear yet, but it may be due to lack of appropriate diagnosis, treatment and cataloging of cancer cases. Because patients seldom exhibit symptoms until an advanced stage of the disease, pancreatic cancer remains one of the most lethal malignant neoplasms that caused 432,242 new deaths in 2018 (GLOBOCAN 2018 estimates). Globally, 458,918 new cases of pancreatic cancer have been reported in 2018, and 355,317 new cases are estimated to occur until 2040. Despite advancements in the detection and management of pancreatic cancer, the 5-year survival rate still stands at 9% only. To date, the causes of pancreatic carcinoma are still insufficiently known, although certain risk factors have been identified, such as tobacco smoking, diabetes mellitus, obesity, dietary factors, alcohol abuse, age, ethnicity, family history and genetic factors, Helicobacter pylori infection, non-O blood group and chronic pancreatitis. In general population, screening of large groups is not considered useful to detect the disease at its early stage, although newer techniques and the screening of tightly targeted groups (especially of those with family history), are being evaluated. Primary prevention is considered of utmost importance. Up-to-date statistics on pancreatic cancer occurrence and outcome along with a better understanding of the etiology and identifying the causative risk factors are essential for the primary prevention of this disease.
Collapse
Affiliation(s)
- Prashanth Rawla
- Department of Internal Medicine, SOVAH Health, Martinsville, VA 24112, USA
| | - Tagore Sunkara
- Department of Gastroenterology and Hepatology, Mercy Medical Center, Des Moines, IA 50314, USA
| | - Vinaya Gaduputi
- Division of Gastroenterology, SBH Health System, Bronx, NY, USA
| |
Collapse
|
27
|
Roch AM, Schneider J, Carr RA, Lancaster WP, House MG, Zyromski NJ, Nakeeb A, Schmidt CM, Ceppa EP. Are BRCA1 and BRCA2 gene mutation patients underscreened for pancreatic adenocarcinoma? J Surg Oncol 2019; 119:777-783. [PMID: 30636051 DOI: 10.1002/jso.25376] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/30/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Breast cancer (BRCA) mutations account for the highest proportion of hereditary causes of pancreatic ductal adenocarcinoma (PDAC). Screening is currently recommended only for patients with one first-degree relative or two family members with PDAC. We hypothesized that screening all BRCA1/2 patients would identify a higher rate of pancreatic abnormalities. METHODS All BRCA1/2 patients at a single academic center were retrospectively reviewed (2005-2015). Pancreatic abnormalities were defined on cross-sectional imaging as pancreatic neoplasm (cystic/solid) or main-duct dilation. RESULTS Two hundred and four patients were identified with BRCA mutations. Forty-seven (40%) had abdominal imaging (20 computerized tomography and 27 magnetic resonance imaging). Twenty-one percent had pancreatic abnormalities (PDAC [n = 2] and intraductal papillary mucinous neoplasm [IPMN; n = 8]). The prevalence of pancreatic abnormalities and IPMN was higher in BRCA2 patients than in the general population (21% vs 8% and 17% vs 1%; P = 0.0007 and P < 0.0001, respectively), with no influence of family history. Similarly, BRCA1 patients had an increased prevalence of IPMN (8.3% vs 1%; P < 0.0001). CONCLUSIONS In this series, 4% and 17% of BRCA2 patients developed PDAC and IPMN, respectively. Eight percent of BRCA1 patients developed IPMN. Under current recommended screening, 60% of BRCA1/2 patients had incompletely pancreatic assessment. With no influence of family history, this study suggests all BRCA1/2 patients should undergo a high-risk screening protocol that will identify a higher rate of precancerous pancreatic neoplasms amenable to curative resection.
Collapse
Affiliation(s)
- Alexandra M Roch
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Justine Schneider
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Rosalie A Carr
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - William P Lancaster
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Michael G House
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Nicholas J Zyromski
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Attila Nakeeb
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - C Max Schmidt
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Eugene P Ceppa
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
28
|
Long NP, Yoon SJ, Anh NH, Nghi TD, Lim DK, Hong YJ, Hong SS, Kwon SW. A systematic review on metabolomics-based diagnostic biomarker discovery and validation in pancreatic cancer. Metabolomics 2018; 14:109. [PMID: 30830397 DOI: 10.1007/s11306-018-1404-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/31/2018] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Metabolomics is an emerging approach for early detection of cancer. Along with the development of metabolomics, high-throughput technologies and statistical learning, the integration of multiple biomarkers has significantly improved clinical diagnosis and management for patients. OBJECTIVES In this study, we conducted a systematic review to examine recent advancements in the oncometabolomics-based diagnostic biomarker discovery and validation in pancreatic cancer. METHODS PubMed, Scopus, and Web of Science were searched for relevant studies published before September 2017. We examined the study designs, the metabolomics approaches, and the reporting methodological quality following PRISMA statement. RESULTS AND CONCLUSION: The included 25 studies primarily focused on the identification rather than the validation of predictive capacity of potential biomarkers. The sample size ranged from 10 to 8760. External validation of the biomarker panels was observed in nine studies. The diagnostic area under the curve ranged from 0.68 to 1.00 (sensitivity: 0.43-1.00, specificity: 0.73-1.00). The effects of patients' bio-parameters on metabolome alterations in a context-dependent manner have not been thoroughly elucidated. The most reported candidates were glutamic acid and histidine in seven studies, and glutamine and isoleucine in five studies, leading to the predominant enrichment of amino acid-related pathways. Notably, 46 metabolites were estimated in at least two studies. Specific challenges and potential pitfalls to provide better insights into future research directions were thoroughly discussed. Our investigation suggests that metabolomics is a robust approach that will improve the diagnostic assessment of pancreatic cancer. Further studies are warranted to validate their validity in multi-clinical settings.
Collapse
Affiliation(s)
- Nguyen Phuoc Long
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Sang Jun Yoon
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Nguyen Hoang Anh
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Tran Diem Nghi
- School of Medicine, Vietnam National University, Ho Chi Minh City, 700000, Vietnam
| | - Dong Kyu Lim
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Yu Jin Hong
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Soon-Sun Hong
- Department of Drug Development, College of Medicine, Inha University, Incheon, 22212, South Korea
| | - Sung Won Kwon
- Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
29
|
Intake of methyl-related nutrients and risk of pancreatic cancer in a population-based case-control study in Minnesota. Eur J Clin Nutr 2018; 72:1128-1135. [PMID: 29904184 PMCID: PMC6119097 DOI: 10.1038/s41430-018-0228-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/17/2018] [Accepted: 05/22/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Folate, vitamin B6, vitamin B12, and methionine are involved in DNA synthesis and methylation and thus may modulate pancreatic cancer risk. We investigated these associations in a population-based case-control study conducted in 1994-1998. SUBJECTS/METHODS Cases (n = 150) were identified from all hospitals in the metropolitan areas of the Twin Cities and the Mayo Clinic, Minnesota. Controls (n = 459) were selected randomly from the general population and were frequency matched to cases by age, sex, and race. Logistic regression was used to estimate odds ratios (OR) and 95% confidence intervals (95% CI) for risk of pancreatic cancer in relation to intake of nutrients considered. RESULTS Dietary intake of folate was associated with a reduced pancreatic cancer risk [OR (95% CI) for quartile (Q) 4 vs. Q1: 0.31 (0.12-0.78)]. A composite score (range from 2 to 8), reflecting combined dietary intake of folate and vitamin B6, was also inversely associated with pancreatic cancer risk [OR (95% CI) for Q4 vs. Q1: 0.24 (0.08-0.70)]. Null associations were found for intake of vitamin B12 and methionine. CONCLUSIONS Dietary folate intake was associated with a reduced pancreatic cancer risk, and this association became stronger when dietary intake of folate and vitamin B6 was combined in analysis.
Collapse
|
30
|
DaVee T, Coronel E, Papafragkakis C, Thaiudom S, Lanke G, Chakinala RC, Nogueras González GM, Bhutani MS, Ross WA, Weston BR, Lee JH. Pancreatic cancer screening in high-risk individuals with germline genetic mutations. Gastrointest Endosc 2018; 87:1443-1450. [PMID: 29309780 DOI: 10.1016/j.gie.2017.12.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/03/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Pancreatic cancer (PC) is a deadly disease that is most commonly diagnosed at an incurable stage. Different high-risk genetic variants and cancer syndromes increase the lifetime risk of developing PC. This study aims to assess the yield of initial PC screening in patients with high-risk germline mutations. METHODS Asymptomatic adults underwent PC screening by EUS, magnetic resonance imaging, or CT during a 10-year period and were retrospectively identified. High-risk individuals were defined as carrying germline mutations in BRCA1, BRCA2, p53 (Li-Fraumeni), STK11 (Peutz-Jeghers), MSH2 (Lynch), ATM (ataxia-telangiectasia), or APC (familial adenomatous polyposis). Patients without germline mutations were excluded. RESULTS In total, 86 patients met the study criteria. The median age was 48.5 years (interquartile range, 40-58), 79.1% (68) were women, and 43.0% (37) had a family history of PC. The genetic mutations were BRCA2 (50, 58.1%), BRCA1 (14, 16.3%), p53 (12, 14.0%), STK11 (5, 5.8%), MSH2 (3, 3.5%), ATM (1, 1.2%), and APC (1, 1.2%). Screening detected a pancreatic abnormality (PA) in 26.7% (23/86), including cysts (11, 47.8%), hyperechoic strands and foci (10, 43.5%), and mild pancreatic duct dilation (2, 8.7%). Patients older than 60 years were more likely to have a PA detected (P = .043). EUS detected more PAs than magnetic resonance imaging or CT. No cases of PC were diagnosed by screening or during follow-up (median, 29.8 months; interquartile range, 21.7-43.5). CONCLUSIONS Unless indicated otherwise by family or personal history, PC screening under the age of 50 is low yield. Linear EUS may be the preferred modality for initial PC screening.
Collapse
Affiliation(s)
- Tomas DaVee
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Emmanuel Coronel
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Charilaos Papafragkakis
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sayam Thaiudom
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gandhi Lanke
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Raja C Chakinala
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Manoop S Bhutani
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - William A Ross
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brian R Weston
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey H Lee
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
31
|
Chen Q, Yuan H, Shi GD, Wu Y, Liu DF, Lin YT, Chen L, Ge WL, Jiang K, Miao Y. Association between NR5A2 and the risk of pancreatic cancer, especially among Caucasians: a meta-analysis of case-control studies. Onco Targets Ther 2018; 11:2709-2723. [PMID: 29785120 PMCID: PMC5953269 DOI: 10.2147/ott.s157759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Previous studies have reported that nuclear receptor subfamily 5, group A, member 2 (NR5A2) polymorphisms (rs3790843 G>A, rs3790844 T>C, rs12029406 C>T) are associated with the risk of pancreatic cancer. However, the results of epidemiological investigations are still controversial. In order to explore its potential attributing factors, we pooled the updated literatures to evaluate the association between NR5A2 polymorphism and the risk of pancreatic cancer in this meta-analysis. Materials and methods Databases such as PubMed, Google Scholar and China National Knowledge Infrastructure were searched for eligible articles following strict inclusion and exclusion criteria (updated to November 18, 2017). Odds ratios (ORs) and 95% CIs were computed to assess the intensity of association. In addition, heterogeneity, sensitivity analysis and publication bias were explored. All statistical analyses were conducted by STATA 14.0. Results Our results showed that the rs3790843 (GA vs GG: OR=0.86, CI=0.76–0.98, P=0.992; GA+AA vs GG: OR=0.83, CI=0.73–0.94, P=0.950; A vs G: OR=0.85, CI=0.78–0.93, P=0.802), rs3790844 (CC vs TT: OR=0.65, CI=0.54–0.78, P=0.617; CC vs TT+CT: OR=0.73, CI=0.62–0.85, P=0.742; C vs T: OR=0.78, CI=0.73–0.84, P=0.555) and rs12029406 (TT vs CC: OR=0.73, CI=0.61–0.89, P=0.483; TT vs CC+CT: OR=0.78, CI=0.66–0.92, P=0.648; T vs C: OR=0.87, CI=0.79–0.95, P=0.837) polymorphisms were associated statistically with the risk of pancreatic cancer. Furthermore, the results of subgroup analysis showed that rs3790843 and rs3790844 polymorphisms were especially related to the risk of pancreatic cancer in Caucasian population. Conclusion Our results revealed that NR5A2 may have a protective effect on pancreatic cancer. However, more well-designed researches are needed to verify the relationship between NR5A2 polymorphisms and the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Qun Chen
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Hao Yuan
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Guo-Dong Shi
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yang Wu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China.,Division of Pancreatic Surgery, Department of General, Visceral, and Transplantation Surgery, Ludwig-Maximilians University, Munich, Germany
| | - Dong-Fang Liu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yu-Ting Lin
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Lei Chen
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Wan-Li Ge
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| | - Yi Miao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Pancreas Institute, Nanjing Medical University, Nanjing, China
| |
Collapse
|
32
|
Camblin AJ, Pace EA, Adams S, Curley MD, Rimkunas V, Nie L, Tan G, Bloom T, Iadevaia S, Baum J, Minx C, Czibere A, Louis CU, Drummond DC, Nielsen UB, Schoeberl B, Pipas JM, Straubinger RM, Askoxylakis V, Lugovskoy AA. Dual Inhibition of IGF-1R and ErbB3 Enhances the Activity of Gemcitabine and Nab-Paclitaxel in Preclinical Models of Pancreatic Cancer. Clin Cancer Res 2018; 24:2873-2885. [DOI: 10.1158/1078-0432.ccr-17-2262] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 01/29/2018] [Accepted: 03/12/2018] [Indexed: 11/16/2022]
|
33
|
Lee HS, Jang CY, Kim SA, Park SB, Jung DE, Kim BO, Kim HY, Chung MJ, Park JY, Bang S, Park SW, Song SY. Combined use of CEMIP and CA 19-9 enhances diagnostic accuracy for pancreatic cancer. Sci Rep 2018; 8:3383. [PMID: 29467409 PMCID: PMC5821821 DOI: 10.1038/s41598-018-21823-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/06/2018] [Indexed: 12/23/2022] Open
Abstract
Carbohydrate antigen (CA) 19-9 is the only diagnostic marker used in pancreatic cancer despite its limitations. Here, we aimed to identify the diagnostic role of CEMIP (also called KIAA1199) combined with CA 19-9 in patients with pancreatic cancer. A retrospective analysis of prospectively collected patient samples was performed to determine the benefit of diagnostic markers in the diagnosis of pancreatic cancer. We investigated CEMIP and CA 19-9 levels in 324 patients with pancreatic cancer and 49 normal controls using serum enzyme-linked immunosorbent assay. Median CA 19-9 and CEMIP levels were 410.5 U/ml (40.8-3342.5) and 0.67 ng/ml (0.40-1.08), respectively, in patients with pancreatic cancer. The AUROC for CA 19-9 and CEMIP were 0.847 (95% confidence interval [CI]: 0.806-0.888) and 0.760 (95% CI: 0.689-0.831), respectively. Combination of CA 19-9 with CEMIP showed markedly improved AUROC over CA 19-9 alone in pancreatic cancer diagnosis (0.94 vs. 0.89; P < 0.0001). CEMIP showed a diagnostic yield of 86.1% (68/79) in CA 19-9 negative pancreatic cancer. Combined use with CEMIP showed significantly improved diagnostic value compared with CA 19-9 alone in pancreatic cancer. Especially, CEMIP may be a complementary marker in pancreatic cancer patients with normal CA 19-9 levels.
Collapse
Affiliation(s)
- Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Chan Young Jang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sun A Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo Been Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Dawoon E Jung
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Bo Ok Kim
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Ha Yan Kim
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Korea
| | - Moon Jae Chung
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Youp Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Woo Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Si Young Song
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
34
|
Zhang D, Xu XL, Li F, Sun HC, Cui YQ, Liu S, Xu PY. Upregulation of the checkpoint protein CHFR is associated with tumor suppression in pancreatic cancers. Oncol Lett 2018; 14:8042-8050. [PMID: 29344247 PMCID: PMC5755226 DOI: 10.3892/ol.2017.7239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 09/27/2017] [Indexed: 12/24/2022] Open
Abstract
The checkpoint with forkhead-associated (FHA) domain and RING-finger (CHFR) protein was identified as a cell cycle checkpoint protein and E3 ubiquitin ligase. In the present study, the potential functions of CHFR in pancreatic cancer were investigated. CHFR expression was measured in five pancreatic cancer cell lines by reverse transcription- quantitative polymerase chain reaction and western blotting. Capan-1 cells stably expressing CHFR were established by lentiviral vector transfection. Cell proliferation was assessed using Cell Counting Kit-8, and cell migration/invasion assay was determined using Transwell assays. Cell cycle and apoptosis induced by gemcitabine or docetaxel were evaluated using flow cytometry. CHFR expression levels were also evaluated in pancreatic ductal adenocarcinoma (PDAC) tumor samples as well as adjacent non-tumor tissues by immunohistochemistry. The significance of CHFR expression was determined, with respect to clinicopathological features and overall survival. Overexpression of CHFR in Capan-1 cells led to a decreased proliferative rate and reduced cell migration and invasion abilities. Results also indicated an increase in G1 phase cells in Capan-1 cells overexpressing CHFR. Docetaxel-induced apoptosis was inhibited in Capan-1 cells with CHFR-overexpression. A reduction in CHFR expression was detected in 51.9% of patients with PDAC, which significantly correlated with later T-stage. The results show CHFR functions as a tumor suppressor in pancreatic cancer, suggests its potential role in controlling the cell cycle of pancreatic cancer cells; however, CHFR overexpression is not a favorable factor in apoptosis induced by docetaxel.
Collapse
Affiliation(s)
- Di Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Xiao-Lan Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Hai-Chen Sun
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Ye-Qing Cui
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Shuang Liu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Ping-Yong Xu
- National Key Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
| |
Collapse
|
35
|
Abstract
Pancreatic cancer is a highly aggressive malignant disease having very limited therapeutic options that ultimately results in its poor prognosis. It is still elusive on the etiology and tumorigenic mechanisms of pancreatic cancer. In the present report, we provide evidence showing involvement of the mineral dust-induced gene (mdig) in the pathogenesis and prognosis of the pancreatic cancer. Using immunohistochemistry approach on human pancreatic cancer tissue microarray, we found differential expression of mdig in pancreatic adenocarcinoma and normal pancreas. Based on the staining intensities of mdig in these tissue samples, we found that 12% of the cancer tissues were strongly positive for mdig, 39% and 31% were moderately and weakly positive respectively. Several alternatively spliced mdig mRNAs were detected in the selected pancreatic cancer cell lines. Through R2 platform for the patient survival analysis (http://r2.amc.nl), we found that enrichment of some specific exon of mdig predicates different survival rate of the pancreatic cancer patients. In summary, our findings may help in assessing the role of mdig in the pathogenesis of the pancreatic cancer and the prognosis of the pancreatic cancer patients.
Collapse
Affiliation(s)
- Srinivas Ashok Kumar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Chitra Thakur
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Lingzhi Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, The Institute of Sericulture and Systems Biology, Southwest University, Chongqing, China
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
36
|
Chen J, Domingue JC, Sears CL. Microbiota dysbiosis in select human cancers: Evidence of association and causality. Semin Immunol 2017; 32:25-34. [PMID: 28822617 DOI: 10.1016/j.smim.2017.08.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/25/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023]
Abstract
The human microbiota is a complex ecosystem of diverse microorganisms consisting of bacteria, viruses, and fungi residing predominantly in epidermal and mucosal habitats across the body, such as skin, oral cavity, lung, intestine and vagina. These symbiotic communities in health, or dysbiotic communities in disease, display tremendous interaction with the local environment and systemic responses, playing a critical role in the host's nutrition, immunity, metabolism and diseases including cancers. While the profiling of normal microbiota in healthy populations is useful and necessary, more recent studies have focused on the microbiota associated with disease, particularly cancers. In this paper, we review current evidence on the role of the human microbiota in four cancer types (colorectal cancer, head and neck cancer, pancreatic cancer, and lung cancer) proposed as affected by both the oral and gut microbiota, and provide a perspective on current gaps in the knowledge of the microbiota and cancer.
Collapse
Affiliation(s)
- Jie Chen
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Jada C Domingue
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Cynthia L Sears
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA; Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, Division of Tumor Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Bloomberg-Kimmel Cancer Immunotherapy Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
37
|
Moutinho-Ribeiro P, Coelho R, Giovannini M, Macedo G. Pancreatic cancer screening: Still a delusion? Pancreatology 2017; 17:754-765. [PMID: 28739291 DOI: 10.1016/j.pan.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/17/2017] [Accepted: 07/05/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic adenocarcinoma represents the fourth most common cause of cancer mortality and death due to pancreatic cancer (PC) have increased since 2003. Its incidence has also raised about 30% in the past decade and it is expected to become the second cause of cancer mortality by 2020 in the USA. Most PC present with metastatic disease and improvements in treatment outcomes for this group have been disappointing. These observations support the idea that screening to identify patients at an earlier stage might be an important strategy in improving overall PC outcomes. Many protocols have been tested, nevertheless, by now there is no effective screening program. Given the overall low incidence of disease and the current lack of accurate, inexpensive and noninvasive screening tests, the consensus is that widespread population-based screening for PC in the general population or in patients with only one affected first-degree relative is neither practicable nor indicated in most countries. However, a different scenario is screening patients with higher risk for PC, most of them with hereditary conditions predisposing the development of this neoplasia. In fact, some guidelines are now available helping to select these individuals at risk and to screen them, in order to achieve early detection of PC.
Collapse
Affiliation(s)
- Pedro Moutinho-Ribeiro
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal.
| | - Rosa Coelho
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal
| | - Marc Giovannini
- Endoscopic Unit, Paoli-Calmettes Institute, Marseilles, France
| | - Guilherme Macedo
- Department of Gastroenterology, Centro Hospitalar São João, Porto, Portugal; Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|
38
|
McAllister F, Montiel MF, Uberoi GS, Uberoi AS, Maitra A, Bhutani MS. Current Status and Future Directions for Screening Patients at High Risk for Pancreatic Cancer. Gastroenterol Hepatol (N Y) 2017; 13:268-275. [PMID: 28656024 PMCID: PMC5479340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
It is well known that pancreatic ductal adenocarcinoma has a high mortality rate. Despite progress in understanding the biology and genetic basis of this disease, life expectancy has changed minimally in the last 50 years. This article highlights the importance of screening patients at high risk for developing pancreatic cancer and reviews current methods as well as methods in development for pancreatic cancer early detection and surveillance.
Collapse
Affiliation(s)
- Florencia McAllister
- Dr McAllister is an assistant professor and Dr Montiel is a research investigator in the Department of Clinical Cancer Prevention; Dr Guneesh S. Uberoi and Dr Angad S. Uberoi are research interns and Dr Bhutani is a professor in the Department of Gastroenterology, Hepatology, and Nutrition; and Dr Maitra is a professor in the Department of Pathology at the University of Texas MD Anderson Cancer Center in Houston, Texas
| | - Maria F Montiel
- Dr McAllister is an assistant professor and Dr Montiel is a research investigator in the Department of Clinical Cancer Prevention; Dr Guneesh S. Uberoi and Dr Angad S. Uberoi are research interns and Dr Bhutani is a professor in the Department of Gastroenterology, Hepatology, and Nutrition; and Dr Maitra is a professor in the Department of Pathology at the University of Texas MD Anderson Cancer Center in Houston, Texas
| | - Guneesh S Uberoi
- Dr McAllister is an assistant professor and Dr Montiel is a research investigator in the Department of Clinical Cancer Prevention; Dr Guneesh S. Uberoi and Dr Angad S. Uberoi are research interns and Dr Bhutani is a professor in the Department of Gastroenterology, Hepatology, and Nutrition; and Dr Maitra is a professor in the Department of Pathology at the University of Texas MD Anderson Cancer Center in Houston, Texas
| | - Angad S Uberoi
- Dr McAllister is an assistant professor and Dr Montiel is a research investigator in the Department of Clinical Cancer Prevention; Dr Guneesh S. Uberoi and Dr Angad S. Uberoi are research interns and Dr Bhutani is a professor in the Department of Gastroenterology, Hepatology, and Nutrition; and Dr Maitra is a professor in the Department of Pathology at the University of Texas MD Anderson Cancer Center in Houston, Texas
| | - Anirban Maitra
- Dr McAllister is an assistant professor and Dr Montiel is a research investigator in the Department of Clinical Cancer Prevention; Dr Guneesh S. Uberoi and Dr Angad S. Uberoi are research interns and Dr Bhutani is a professor in the Department of Gastroenterology, Hepatology, and Nutrition; and Dr Maitra is a professor in the Department of Pathology at the University of Texas MD Anderson Cancer Center in Houston, Texas
| | - Manoop S Bhutani
- Dr McAllister is an assistant professor and Dr Montiel is a research investigator in the Department of Clinical Cancer Prevention; Dr Guneesh S. Uberoi and Dr Angad S. Uberoi are research interns and Dr Bhutani is a professor in the Department of Gastroenterology, Hepatology, and Nutrition; and Dr Maitra is a professor in the Department of Pathology at the University of Texas MD Anderson Cancer Center in Houston, Texas
| |
Collapse
|
39
|
Lower plasma levels of glucose-dependent insulinotropic peptide (GIP) and pancreatic polypeptide (PP) in patients with ductal adenocarcinoma of the pancreas and their relation to the presence of impaired glucoregulation and weight loss. Pancreatology 2016; 17:89-94. [PMID: 28027898 DOI: 10.1016/j.pan.2016.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Revised: 11/14/2016] [Accepted: 12/15/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND The changes in gastrointestinal hormones associated with pancreatic ductal adenocarcinoma (PDAC) in patients with impaired glucoregulation have yet to be evaluated. The aim of this study was to determine plasma concentrations of selected gastrointestinal hormones in PDAC patients with and without diabetes and to compare them with levels found in Type 2 diabetic patients without cancer. METHODS In this study we examined plasma concentrations of glucose-dependent insulinotropic peptide (GIP), glucagon-like peptide 1 (GLP-1), pancreatic polypeptide (PP), peptide YY (PYY) and neuropeptide Y (NPY), and cytokines leptin and adiponectin in 94 patients with histologically confirmed PDAC. Thirty-nine patients with Type 2 diabetes without PDAC and 29 healthy individuals with no evidence of acute or chronic diseases were examined as controls. RESULTS Significantly lower plasma concentrations of GIP were found in PDAC patients with new-onset diabetes/prediabetes (n = 76), or in those with normal glucose regulation (n = 18), compared to patients with Type 2 diabetes without PDAC and controls (15.5 (3.7-64.5) or 6.5 (1.7-24.5) vs. 39.8 (15.1-104.7) and 28.8 (7.4-112.2) ng/L, p < 0.001); the same relationship was observed for PP (38.9 (10.2-147.9) or 28.1 (7.9-100.0) vs 89.1 (38.0-208.9) and 75.8 (30.1-190.6) ng/L, p < 0.01), respectively. The lowest levels of GIP and PP concentrations were found in PDAC patients with new-onset diabetes/prediabetes and weight loss > 2 kg (p < 0.001). CONCLUSIONS We conclude that GIP and PP plasma concentrations are lower in pancreatic cancer irrespective of the degree of glucose intolerance as compared to Type 2 diabetic patients and healthy controls. In new onset diabetes especially if associated with weight loss, these changes may represent a new clue for the diagnosis of PDAC.
Collapse
|
40
|
Early detection of pancreatic cancer: impact of high-resolution imaging methods and biomarkers. Eur J Gastroenterol Hepatol 2016; 28:e33-e43. [PMID: 27769077 DOI: 10.1097/meg.0000000000000727] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
High-resolution imaging methods (HRIMs) and biomarkers present the second step of pancreatic cancer (PC) diagnostics in at-risk individuals. These include patients with positive risk factors, early symptoms, nonresponders to the initial antidiabetic therapy, patients older than 50 years of age with new-onset unstable diabetes requiring insulin as well as patients with long-term insulin-non-dependent diabetes and recent (up to 6 months) failure of antidiabetic therapy. The procedures should be started without delay and the co-operation between the primary and tertiary medical centers is highly desirable. An early indication of HRIMs and biomarkers is a prerequisite for the diagnosis of a resectable PC. This publication reviews the recent contribution of HRIMs and biomarkers toward an early diagnosis of PC.
Collapse
|
41
|
Ilic M, Ilic I. Epidemiology of pancreatic cancer. World J Gastroenterol 2016; 22:9694-9705. [PMID: 27956793 PMCID: PMC5124974 DOI: 10.3748/wjg.v22.i44.9694] [Citation(s) in RCA: 939] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 08/30/2016] [Accepted: 09/28/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer of the pancreas remains one of the deadliest cancer types. Based on the GLOBOCAN 2012 estimates, pancreatic cancer causes more than 331000 deaths per year, ranking as the seventh leading cause of cancer death in both sexes together. Globally, about 338000 people had pancreatic cancer in 2012, making it the 11th most common cancer. The highest incidence and mortality rates of pancreatic cancer are found in developed countries. Trends for pancreatic cancer incidence and mortality varied considerably in the world. A known cause of pancreatic cancer is tobacco smoking. This risk factor is likely to explain some of the international variations and gender differences. The overall five-year survival rate is about 6% (ranges from 2% to 9%), but this vary very small between developed and developing countries. To date, the causes of pancreatic cancer are still insufficiently known, although certain risk factors have been identified, such as smoking, obesity, genetics, diabetes, diet, inactivity. There are no current screening recommendations for pancreatic cancer, so primary prevention is of utmost importance. A better understanding of the etiology and identifying the risk factors is essential for the primary prevention of this disease.
Collapse
|
42
|
Lee KH, Dominici F, Schrag D, Haneuse S. Hierarchical models for semi-competing risks data with application to quality of end-of-life care for pancreatic cancer. J Am Stat Assoc 2016; 111:1075-1095. [PMID: 28303074 DOI: 10.1080/01621459.2016.1164052] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Readmission following discharge from an initial hospitalization is a key marker of quality of health care in the United States. For the most part, readmission has been studied among patients with 'acute' health conditions, such as pneumonia and heart failure, with analyses based on a logistic-Normal generalized linear mixed model (Normand et al., 1997). Naïve application of this model to the study of readmission among patients with 'advanced' health conditions such as pancreatic cancer, however, is problematic because it ignores death as a competing risk. A more appropriate analysis is to imbed such a study within the semi-competing risks framework. To our knowledge, however, no comprehensive statistical methods have been developed for cluster-correlated semi-competing risks data. To resolve this gap in the literature we propose a novel hierarchical modeling framework for the analysis of cluster-correlated semi-competing risks data that permits parametric or non-parametric specifications for a range of components giving analysts substantial flexibility as they consider their own analyses. Estimation and inference is performed within the Bayesian paradigm since it facilitates the straightforward characterization of (posterior) uncertainty for all model parameters, including hospital-specific random effects. Model comparison and choice is performed via the deviance information criterion and the log-pseudo marginal likelihood statistic, both of which are based on a partially marginalized likelihood. An efficient computational scheme, based on the Metropolis-Hastings-Green algorithm, is developed and had been implemented in the SemiCompRisks R package. A comprehensive simulation study shows that the proposed framework performs very well in a range of data scenarios, and outperforms competitor analysis strategies. The proposed framework is motivated by and illustrated with an on-going study of the risk of readmission among Medicare beneficiaries diagnosed with pancreatic cancer. Using data on n=5,298 patients at J=112 hospitals in the six New England states between 2000-2009, key scientific questions we consider include the role of patient-level risk factors on the risk of readmission and the extent of variation in risk across hospitals not explained by differences in patient case-mix.
Collapse
Affiliation(s)
- Kyu Ha Lee
- Epidemiology and Biostatistics Core, The Forsyth Institute, Department of Oral Health Policy and Epidemiology, Harvard School of Dental Medicine
| | | | - Deborah Schrag
- Department of Medical Oncology, Dana Farber Cancer Institute
| | - Sebastien Haneuse
- Department of Biostatistics, Harvard T.H. Chan School of Public Health
| |
Collapse
|
43
|
Inpatient Burden of Pancreatic Cancer in the United States: An Analysis of National Trends in the United States From 1997 to 2012. Pancreas 2016; 45:e41-2. [PMID: 27518364 DOI: 10.1097/mpa.0000000000000648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
44
|
Eloubeidi MA, Decker GA, Chandrasekhara V, Chathadi KV, Early DS, Evans JA, Fanelli RD, Fisher DA, Foley K, Hwang JH, Jue TL, Lightdale JR, Pasha SF, Saltzman JR, Sharaf R, Shergill AK, Cash BD, DeWitt JM. The role of endoscopy in the evaluation and management of patients with solid pancreatic neoplasia. Gastrointest Endosc 2016; 83:17-28. [PMID: 26706297 DOI: 10.1016/j.gie.2015.09.009] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023]
|
45
|
Tan XP, Dong WG, Zhang Q, Yang ZR, Lei XF, Ai MH. Potential effects of Mina53 on tumor growth in human pancreatic cancer. Cell Biochem Biophys 2015; 69:619-25. [PMID: 24522517 PMCID: PMC4057634 DOI: 10.1007/s12013-014-9841-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Myc-induced nuclear antigen (Mina53) is a protein with a molecular weight of 53 kDa expression of which is induced by c-Myc. Increased expression of Mina53 is documented in some human carcinomas. In this study, we found markedly increased Mina53 expression in pancreatic cancer tissue specimens. This expression did not correlate with clinicopathological characteristics, such as sex, age, and presence of distant metastasis. However, there was a statistically significant association with histological differentiation, TNM stage, and lymph node metastases. To study functional role of Mina53, we silenced its expression by siRNA in PANC-1 cells. These cells were arrested in the G2/M phase, and apoptosis rates were increased. In conclusion, increased expression of Mina53 may play an important role in the development of human pancreatic cancer. Mina53 can be used as a marker for pancreatic cancer and may potentially be exploited as a target for treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Xiao-ping Tan
- Department of Gastroenterology, No. 1 Hospital, Yangtze University, Jingzhou, 434000, Hubei, People's Republic of China,
| | | | | | | | | | | |
Collapse
|
46
|
Rahman F, Cotterchio M, Cleary SP, Gallinger S. Association between alcohol consumption and pancreatic cancer risk: a case-control study. PLoS One 2015; 10:e0124489. [PMID: 25856529 PMCID: PMC4391718 DOI: 10.1371/journal.pone.0124489] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/03/2015] [Indexed: 12/15/2022] Open
Abstract
Purpose Evidence is inconsistent regarding alcohol and pancreatic cancer risk, although heavy drinking may increase risk. Methods A population-based case-control study was conducted using 345 pancreas cancer cases diagnosed 2011–2012 and 1,285 frequency-matched controls from Ontario, Canada. Logistic regression was used to evaluate alcohol consumption and pancreatic cancer risk; data was also stratified by sex and smoking status to assess interaction. Results Alcohol consumption was not associated with pancreatic cancer risk (age-adjusted odds ratio=0.78, 95% CI: 0.58, 1.05 for 1 - 3 drinks/week; age-adjusted odds ratio=0.86, 95% CI: 0.63, 1.17 for 4 - 20 drinks/week), however there was a non-significant increased risk for heavy drinkers consuming ≥21 drinks/week (age-adjusted odds ratio=1.35, 95% CI: 0.81, 2.27). Cigarette smoking modified the alcohol-cancer relationship; among current smokers, heavy alcohol consumption was associated with a significantly increased pancreatic cancer risk (age-adjusted odds ratio=4.04, 95% CI: 1.58, 10.37), whereas this significant association with heavy drinking was not observed among non-smokers (age-adjusted odds ratio=2.01, 95% CI: 0.50, 8.18). Furthermore, light – moderate alcohol intake was associated with increased pancreas cancer risk among current smokers. Conclusions While alcohol was not significantly associated with pancreatic cancer risk, smoking status modified this relationship such that among current smokers, alcohol intake was associated with a greater than two-fold increased risk of pancreatic cancer. The results should be interpreted with caution due to small sample sizes within subgroups and correction for multiple comparisons should be considered. These findings should be replicated in larger studies where more precise estimates of risk can be obtained.
Collapse
Affiliation(s)
- Farah Rahman
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| | - Michelle Cotterchio
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Prevention and Cancer Control, Cancer Care Ontario, Toronto, Ontario, Canada
| | - Sean P. Cleary
- Department of Surgery, University Health Network, Toronto, Ontario, Canada
- Prosserman Centre for Health Research, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of General Surgery, Toronto General Hospital, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Pandharipande PV, Heberle C, Dowling EC, Kong CY, Tramontano A, Perzan KE, Brugge W, Hur C. Targeted screening of individuals at high risk for pancreatic cancer: results of a simulation model. Radiology 2015; 275:177-87. [PMID: 25393849 PMCID: PMC4372492 DOI: 10.1148/radiol.14141282] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE To identify when, from the standpoint of relative risk, magnetic resonance (MR) imaging-based screening may be effective in patients with a known or suspected genetic predisposition to pancreatic cancer. MATERIALS AND METHODS The authors developed a Markov model of pancreatic ductal adenocarcinoma (PDAC). The model was calibrated to National Cancer Institute Surveillance, Epidemiology, and End Results registry data and informed by the literature. A hypothetical screening strategy was evaluated in which all population individuals underwent one-time MR imaging screening at age 50 years. Screening outcomes for individuals with an average risk for PDAC ("base case") were compared with those for individuals at an increased risk to assess for differential benefits in populations with a known or suspected genetic predisposition. Effects of varying key inputs, including MR imaging performance, surgical mortality, and screening age, were evaluated with a sensitivity analysis. RESULTS In the base case, screening resulted in a small number of cancer deaths averted (39 of 100 000 men, 38 of 100 000 women) and a net decrease in life expectancy (-3 days for men, -4 days for women), which was driven by unnecessary pancreatic surgeries associated with false-positive results. Life expectancy gains were achieved if an individual's risk for PDAC exceeded 2.4 (men) or 2.7 (women) times that of the general population. When relative risk increased further, for example to 30 times that of the general population, averted cancer deaths and life expectancy gains increased substantially (1219 of 100 000 men, life expectancy gain: 65 days; 1204 of 100 000 women, life expectancy gain: 71 days). In addition, results were sensitive to MR imaging specificity and the surgical mortality rate. CONCLUSION Although PDAC screening with MR imaging for the entire population is not effective, individuals with even modestly increased risk may benefit.
Collapse
Affiliation(s)
- Pari V. Pandharipande
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - Curtis Heberle
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - Emily C. Dowling
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - Chung Yin Kong
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - Angela Tramontano
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - Katherine E. Perzan
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - William Brugge
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| | - Chin Hur
- From the Massachusetts General Hospital Institute for Technology
Assessment (P.V.P., C.H., E.C.D., C.Y.K., A.T., K.E.P., C.H.), Department of
Radiology (P.V.P., C.H., E.C.D., C.Y.K., A.T.), and Department of General Medicine,
Gastrointestinal Unit (K.E.P., W.B., C.H.), Massachusetts General Hospital, 101
Merrimac St, 10th Floor, Boston, MA 02114; and Harvard Medical School, Boston,
Mass
| |
Collapse
|
48
|
Abstract
Only 30% of patients with a diagnosis of pancreatic cancer survive 1 year after the diagnosis. Progress in understanding the causes of pancreatic cancer has been made, including solidifying the associations with obesity and diabetes, and a proportion of cases should be preventable through lifestyle modifications. Unfortunately, identifying reliable biomarkers of early pancreatic cancer has been extremely challenging, and no effective screening modality is currently available for this devastating form of cancer. Recent data suggest that the microbiota may play a role in the disease process, but many questions remain. Future studies focusing on the human microbiome, both etiologically and as a marker of disease susceptibility, should shed light on how to better tackle prevention, early detection, and treatment of this highly fatal disease.
Collapse
Affiliation(s)
- Dominique S Michaud
- From the *Department of Epidemiology, School of Public Health, Brown University, Providence, RI; †Department of Epidemiology and Public Health, Imperial College, London, United Kingdom; ‡The Forsyth Institute, Cambridge MA; and §Harvard School of Dental Medicine, Boston MA
| | | |
Collapse
|
49
|
Familial pancreatic cancer: the case for prophylactic pancreatectomy in lieu of serial screening and shared decision making. Case Rep Oncol Med 2014; 2014:737183. [PMID: 25506012 PMCID: PMC4258333 DOI: 10.1155/2014/737183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/10/2014] [Accepted: 11/13/2014] [Indexed: 01/02/2023] Open
Abstract
At-risk family members with familial pancreatic cancer (FCaP) face uncertainty regarding the individual risk of developing pancreatic cancer (CaP) and whether to choose serial screening or prophylactic pancreatectomy to avoid CaP. We treated 2 at-risk siblings with a history of FCaP, congenital hepatic fibrosis (CHF), and jaundice secondary to a bile duct stricture. In one, a pancreaticoduodenal resection was done and in the second a total pancreatectomy. Malignancy was not present, but extensive pancreatic intraepithelial neoplasia (PanIn) 2 was present throughout both pancreata. The clinical course and literature review are presented along with the previously unreported association of CHF and CaP.
Collapse
|
50
|
Lennon AM, Victor D, Zaheer A, Ostovaneh MR, Jeh J, Law JK, Rezaee N, Molin MD, Ahn YJ, Wu W, Khashab MA, Girotra M, Ahuja N, Makary MA, Weiss MJ, Hirose K, Goggins M, Hruban RH, Cameron A, Wolfgang CL, Singh VK, Gurakar A. Liver transplant patients have a risk of progression similar to that of sporadic patients with branch duct intraductal papillary mucinous neoplasms. Liver Transpl 2014; 20:1462-7. [PMID: 25155689 PMCID: PMC4322915 DOI: 10.1002/lt.23983] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 08/08/2014] [Accepted: 08/17/2014] [Indexed: 12/21/2022]
Abstract
Intraductal papillary mucinous neoplasms (IPMNs) have malignant potential and can progress from low- to high-grade dysplasia to invasive adenocarcinoma. The management of patients with IPMNs is dependent on their risk of malignant progression, with surgical resection recommended for patients with branch-duct IPMN (BD-IPMN) who develop high-risk features. There is increasing evidence that liver transplant (LT) patients are at increased risk of extrahepatic malignancy. However, there are few data regarding the risk of progression of BD-IPMNs in LT recipients. The aim of this study was to determine whether LT recipients with BD-IPMNs are at higher risk of developing high-risk features than patients with BD-IPMNs who did not receive a transplant. Consecutive patients who underwent an LT with BD-IPMNs were included. Patients with BD-IPMNs with no history of immunosuppression were used as controls. Progression of the BD-IPMNs was defined as development of a high-risk feature (jaundice, dilated main pancreatic duct, mural nodule, cytology suspicious or diagnostic for malignancy, cyst diameter ≥3 cm). Twenty-three LT patients with BD-IPMN were compared with 274 control patients. The median length of follow-up was 53.7 and 24.0 months in LT and control groups, respectively. Four (17.4%) LT patients and 45 (16.4%) controls developed high-risk features (P = 0.99). In multivariate analysis, progression of BD-IPMNs was associated with age at diagnosis but not with LT. There was no statistically significant difference in the risk of developing high-risk features between the LT and the control groups.
Collapse
Affiliation(s)
- Anne Marie Lennon
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD,Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - David Victor
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Atif Zaheer
- Department of Radiology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mohammad Reza Ostovaneh
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Jessica Jeh
- Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Joanna K. Law
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Neda Rezaee
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Marco Dal Molin
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Young Joon Ahn
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Wenchuan Wu
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mouen A. Khashab
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Mohit Girotra
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Nita Ahuja
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Martin A. Makary
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Matthew J. Weiss
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Kenzo Hirose
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Michael Goggins
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD,Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Andrew Cameron
- Division of Surgery, The Johns Hopkins Medical Institutions, Baltimore, MD
| | | | - Vikesh K. Singh
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| | - Ahmet Gurakar
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|