1
|
Pian LL, Song MH, Wang TF, Qi L, Peng TL, Xie KP. Identification and analysis of pancreatic intraepithelial neoplasia: opportunities and challenges. Front Endocrinol (Lausanne) 2025; 15:1401829. [PMID: 39839479 PMCID: PMC11746065 DOI: 10.3389/fendo.2024.1401829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Pancreatic intraepithelial neoplasia (PanIN) is the most common precursor lesion of pancreatic ductal adenocarcinoma (PDAC), which has poor prognosis with a short median overall survival of 6-12 months and a low 5-year survival rate of approximately 3%. It is crucial to remove PanIN lesions to prevent the development of invasive PDAC, as PDAC spreads rapidly outside the pancreas. This review aims to provide the latest knowledge on PanIN risk, pathology, cellular origin, genetic susceptibility, and diagnosis, while identifying research gaps that require further investigation in this understudied area of precancerous lesions. PanINs are classified into PanIN 1, PanIN 2, and PanIN 3, with PanIN 3 having the highest likelihood of developing into invasive PDAC. Differentiating between PanIN 2 and PanIN 3 is clinically significant. Genetic alterations found in PDAC are also present in PanIN and increase with the grade of PanIN. Imaging methods alone are insufficient for distinguishing PanIN, necessitating the use of genetic and molecular tests for identification. In addition, metabolomics technologies and miRNAs are playing an increasingly important role in the field of cancer diagnosis, offering more possibilities for efficient identification of PanIN. Although detecting and stratifying the risk of PanIN poses challenges, the combined utilization of imaging, genetics, and metabolomics holds promise for improving patient survival in this field.
Collapse
Affiliation(s)
- Ling-ling Pian
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Mei-hui Song
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Teng-fei Wang
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Tie-li Peng
- Division of Gastroenterology, Institute of Digestive Disease, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, Guangdong, China
| | - Ke-ping Xie
- School of Medicine, The South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Park MA, Gumpper-Fedus K, Krishna SG, Genilo-Delgado MC, Brantley S, Hart PA, Dillhoff ME, Gomez MF, Basinski TL, Mok SR, Luthra AK, Fleming JB, Mohammadi A, Centeno BA, Jiang K, Karolak A, Jeong D, Chen DT, Stewart PA, Teer JK, Cruz-Monserrate Z, Permuth JB. Molecular Pathway and Immune Profile Analysis of IPMN-Derived Versus PanIN-Derived Pancreatic Ductal Adenocarcinomas. Int J Mol Sci 2024; 25:13164. [PMID: 39684873 PMCID: PMC11642437 DOI: 10.3390/ijms252313164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is a dire clinical need. Therefore, we aimed to identify molecular signatures that distinguished between PDAC with and without clinical evidence of an IPMN to identify novel molecular pathways related to IPMN-derived PDAC that could help guide biomarker development. Data from the Oncology Research Information Exchange Network (ORIEN) multi-institute sequencing project were utilized to analyze 66 PDAC cases from Moffitt Cancer Center and The Ohio State University Wexner Medical Center, for which tumor whole transcriptome sequencing datasets were generated. Cases were classified based on whether a tumor had originated from an IPMN (n = 16) or presumably through the pancreatic intraepithelial neoplasia (PanIN) pathway (n = 50). We then performed differential expression and pathway analysis using Gene-Set Enrichment Analysis (GSEA) and Pathway Analysis with Down-weighted Genes (PADOG) algorithms. We also analyzed immune profiles using the Tumor-Immune Microenvironment Deconvolution web portal for Bulk Transcriptomics (TIMEx). Both GSEA and TIMEx indicate that PanIN-derived PDAC tumors enrich inflammatory pathways (complement, hedgehog signaling, coagulation, inflammatory response, apical surface, IL-2/STAT5, IL-6/STAT3, EMT, KRAS signaling, apical junction, IFN-gamma, allograft rejection) and are comparatively richer in almost all immune cell types than those from IPMN-derived PDAC. IPMN-derived tumors were enriched for metabolic and energy-generating pathways (oxidative phosphorylation, unfolded protein response, pancreas beta cells, adipogenesis, fatty acid metabolism, protein secretion), and the most significantly upregulated genes (padj < 0.001) included mucin 2 (MUC2) and gastrokine-2 (GKN2). Further, the metabolic-linked gene signature enriched in the IPMN-derived samples is associated with a cluster of early-stage and long-survival (top 4th quartile) PDAC cases from The Cancer Genome Atlas (TCGA) expression database. Our data suggest that IPMN-derived and PanIN-derived PDACs differ in the expression of immune profiles and metabolic pathways. These initial findings warrant validation and follow-up to develop biomarker-based strategies for early PDAC detection and treatment.
Collapse
Affiliation(s)
- Margaret A. Park
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.-T.C.); (P.A.S.); (J.K.T.)
| | - Kristyn Gumpper-Fedus
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (K.G.-F.); (S.G.K.); (P.A.H.)
| | - Somashekar G. Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (K.G.-F.); (S.G.K.); (P.A.H.)
| | - Maria C. Genilo-Delgado
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
| | - Stephen Brantley
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (S.B.); (B.A.C.); (K.J.)
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (K.G.-F.); (S.G.K.); (P.A.H.)
| | - Mary E. Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA;
| | - Maria F. Gomez
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Toni L. Basinski
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
| | - Shaffer R. Mok
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
| | - Anjuli K. Luthra
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
| | - Jason B. Fleming
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Amir Mohammadi
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
| | - Barbara A. Centeno
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (S.B.); (B.A.C.); (K.J.)
| | - Kun Jiang
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (S.B.); (B.A.C.); (K.J.)
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Daniel Jeong
- Department of Radiology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.-T.C.); (P.A.S.); (J.K.T.)
| | - Paul A. Stewart
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.-T.C.); (P.A.S.); (J.K.T.)
| | - Jamie K. Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA; (D.-T.C.); (P.A.S.); (J.K.T.)
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (K.G.-F.); (S.G.K.); (P.A.H.)
| | - Jennifer B. Permuth
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (M.A.P.); (M.C.G.-D.); (M.F.G.); (T.L.B.); (S.R.M.); (A.K.L.); (A.M.)
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
3
|
Tavano F, Latiano A, Palmieri O, Gioffreda D, Latiano T, Gentile A, Tardio M, Latiano TP, Gentile M, Terracciano F, Perri F. Duodenal Fluid Analysis as a Rewarding Approach to Detect Low-Abundance Mutations in Biliopancreatic Cancers. Int J Mol Sci 2024; 25:8436. [PMID: 39126005 PMCID: PMC11312909 DOI: 10.3390/ijms25158436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Diagnosis of biliopancreatic cancers by the available serum tumor markers, imaging, and histopathological tissue specimen examination remains a challenge. Circulating cell-free DNA derived from matched pairs of secretin-stimulated duodenal fluid (DF) and plasma from 10 patients with biliopancreatic diseases and 8 control subjects was analyzed using AmpliSeq™ HD technology for Ion Torrent Next-Generation Sequencing to evaluate the potential of liquid biopsy with DF in biliopancreatic cancers. The median cfDNA concentration was greater in DF-derived than in plasma-derived samples. A total of 13 variants were detected: 11 vs. 1 were exclusive for DF relative to the plasma source, and 1 was shared between the two body fluids. According to the four-tier systems, 10 clinical tier-I-II (76.9%), 1 tier-III (7.7%), and 2 tier-IV (15.4%) variants were identified. Notably, the 11 tier-I-III variants were exclusively found in DF-derived cfDNA from five patients with biliopancreatic cancers, and were detected in seven genes (KRAS, TP53, BRAF, CDKN2A, RNF43, GNAS, and PIK3CA); 82% of the tier-I-III variants had a low abundance, with a VAF < 6%. The mutational profiling of DF seems to be a reliable and promising tool for identifying cancer-associated alterations in malignant cancers of the biliopancreatic tract.
Collapse
Affiliation(s)
- Francesca Tavano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Anna Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Orazio Palmieri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Domenica Gioffreda
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Annamaria Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Matteo Tardio
- Department of Surgery, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Tiziana Pia Latiano
- Department of Oncology, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Marco Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Fulvia Terracciano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| | - Francesco Perri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS “Casa Sollievo della Sofferenza” Hospital, Viale Cappuccini 1, 71013 San Giovanni Rotondo, FG, Italy
| |
Collapse
|
4
|
Overbeek KA, Cahen DL, Bruno MJ. The role of endoscopic ultrasound in the detection of pancreatic lesions in high-risk individuals. Fam Cancer 2024; 23:279-293. [PMID: 38573399 PMCID: PMC11255057 DOI: 10.1007/s10689-024-00380-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/17/2024] [Indexed: 04/05/2024]
Abstract
Individuals at high risk of developing pancreatic ductal adenocarcinoma are eligible for surveillance within research programs. These programs employ periodic imaging in the form of magnetic resonance imaging/magnetic resonance cholangiopancreatography or endoscopic ultrasound for the detection of early cancer or high-grade precursor lesions. This narrative review discusses the role of endoscopic ultrasound within these surveillance programs. It details its overall strengths and limitations, yield, burden on patients, and how it compares to magnetic resonance imaging. Finally, recommendations are given when and how to incorporate endoscopic ultrasound in the surveillance of high-risk individuals.
Collapse
Affiliation(s)
- Kasper A Overbeek
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Djuna L Cahen
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marco J Bruno
- Erasmus MC Cancer Institute, Department of Gastroenterology & Hepatology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
5
|
Ungkulpasvich U, Hatakeyama H, Hirotsu T, di Luccio E. Pancreatic Cancer and Detection Methods. Biomedicines 2023; 11:2557. [PMID: 37760999 PMCID: PMC10526344 DOI: 10.3390/biomedicines11092557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/05/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The pancreas is a vital organ with exocrine and endocrine functions. Pancreatitis is an inflammation of the pancreas caused by alcohol consumption and gallstones. This condition can heighten the risk of pancreatic cancer (PC), a challenging disease with a high mortality rate. Genetic and epigenetic factors contribute significantly to PC development, along with other risk factors. Early detection is crucial for improving PC outcomes. Diagnostic methods, including imagining modalities and tissue biopsy, aid in the detection and analysis of PC. In contrast, liquid biopsy (LB) shows promise in early tumor detection by assessing biomarkers in bodily fluids. Understanding the function of the pancreas, associated diseases, risk factors, and available diagnostic methods is essential for effective management and early PC detection. The current clinical examination of PC is challenging due to its asymptomatic early stages and limitations of highly precise diagnostics. Screening is recommended for high-risk populations and individuals with potential benign tumors. Among various PC screening methods, the N-NOSE plus pancreas test stands out with its high AUC of 0.865. Compared to other commercial products, the N-NOSE plus pancreas test offers a cost-effective solution for early detection. However, additional diagnostic tests are required for confirmation. Further research, validation, and the development of non-invasive screening methods and standardized scoring systems are crucial to enhance PC detection and improve patient outcomes. This review outlines the context of pancreatic cancer and the challenges for early detection.
Collapse
Affiliation(s)
| | | | | | - Eric di Luccio
- Hirotsu Bioscience Inc., 22F The New Otani Garden Court, 4-1 Kioi-cho, Chiyoda-ku, Tokyo 102-0094, Japan; (U.U.); (H.H.); (T.H.)
| |
Collapse
|
6
|
Takahashi K, Takeda Y, Ono Y, Isomoto H, Mizukami Y. Current status of molecular diagnostic approaches using liquid biopsy. J Gastroenterol 2023; 58:834-847. [PMID: 37470859 PMCID: PMC10423147 DOI: 10.1007/s00535-023-02024-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/08/2023] [Indexed: 07/21/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers, and developing an efficient and reliable approach for its early-stage diagnosis is urgently needed. Precancerous lesions of PDAC, such as pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasms (IPMN), arise through multiple steps of driver gene alterations in KRAS, TP53, CDKN2A, SMAD4, or GNAS. Hallmark mutations play a role in tumor initiation and progression, and their detection in bodily fluids is crucial for diagnosis. Recently, liquid biopsy has gained attention as an approach to complement pathological diagnosis, and in addition to mutation signatures in cell-free DNA, cell-free RNA, and extracellular vesicles have been investigated as potential diagnostic and prognostic markers. Integrating such molecular information to revise the diagnostic criteria for pancreatic cancer can enable a better understanding of the pathogenesis underlying inter-patient heterogeneity, such as sensitivity to chemotherapy and disease outcomes. This review discusses the current diagnostic approaches and clinical applications of genetic analysis in pancreatic cancer and diagnostic attempts by liquid biopsy and molecular analyses using pancreatic juice, duodenal fluid, and blood samples. Emerging knowledge in the rapidly advancing liquid biopsy field is promising for molecular profiling and diagnosing pancreatic diseases with significant diversity.
Collapse
Affiliation(s)
- Kenji Takahashi
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka Higashi, Asahikawa, Hokkaido, 078-8510, Japan.
| | - Yohei Takeda
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Yusuke Ono
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka Higashi, Asahikawa, Hokkaido, 078-8510, Japan
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| | - Hajime Isomoto
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Tottori, Japan
| | - Yusuke Mizukami
- Division of Metabolism and Biosystemic Science, Gastroenterology, and Hematology/Oncology, Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka Higashi, Asahikawa, Hokkaido, 078-8510, Japan
- Institute of Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| |
Collapse
|
7
|
Systematic review and meta-analysis: Diagnostic performance of DNA alterations in pancreatic juice for the detection of pancreatic cancer. Pancreatology 2022; 22:973-986. [PMID: 35864067 DOI: 10.1016/j.pan.2022.06.260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Pancreatic cancer has a dismal prognosis. So far, imaging has been proven incapable of establishing an early enough diagnosis. Thus, biomarkers are urgently needed for early detection and improved survival. Our aim was to evaluate the pooled diagnostic performance of DNA alterations in pancreatic juice. METHODS A systematic literature search was performed in EMBASE, MEDLINE Ovid, Cochrane CENTRAL and Web of Science for studies concerning the diagnostic performance of DNA alterations in pancreatic juice to differentiate patients with high-grade dysplasia or pancreatic cancer from controls. Study quality was assessed using QUADAS-2. The pooled prevalence, sensitivity, specificity and diagnostic odds ratio were calculated. RESULTS Studies mostly concerned cell-free DNA mutations (32 studies: 939 cases, 1678 controls) and methylation patterns (14 studies: 579 cases, 467 controls). KRAS, TP53, CDKN2A, GNAS and SMAD4 mutations were evaluated most. Of these, TP53 had the highest diagnostic performance with a pooled sensitivity of 42% (95% CI: 31-54%), specificity of 98% (95%-CI: 92%-100%) and diagnostic odds ratio of 36 (95% CI: 9-133). Of DNA methylation patterns, hypermethylation of CDKN2A, NPTX2 and ppENK were studied most. Hypermethylation of NPTX2 performed best with a sensitivity of 39-70% and specificity of 94-100% for distinguishing pancreatic cancer from controls. CONCLUSIONS This meta-analysis shows that, in pancreatic juice, the presence of distinct DNA mutations (TP53, SMAD4 or CDKN2A) and NPTX2 hypermethylation have a high specificity (close to 100%) for the presence of high-grade dysplasia or pancreatic cancer. However, the sensitivity of these DNA alterations is poor to moderate, yet may increase if they are combined in a panel.
Collapse
|
8
|
Hanada K, Shimizu A, Kurihara K, Ikeda M, Yamamoto T, Okuda Y, Tazuma S. Endoscopic approach in the diagnosis of high-grade pancreatic intraepithelial neoplasia. Dig Endosc 2022; 34:927-937. [PMID: 35165942 DOI: 10.1111/den.14240] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/02/2022] [Accepted: 01/16/2022] [Indexed: 02/08/2023]
Abstract
Early diagnosis of pancreatic ductal adenocarcinoma (PDAC) is essential for improving prognosis; however, diagnosing PDAC at an early stage is challenging. In patients with localized high-grade pancreatic intraepithelial neoplasia (HG-PanIN), whose tumorous lesion is undetectable on cross-sectional images such as computed tomography or magnetic resonance image, long-term survival is expected. Pancreatic cystic lesions or main pancreatic duct (MPD) dilatation are important indirect findings for the initial diagnosis of HG-PanIN. Magnetic resonance cholangiopancreatography (MRCP) and endoscopic ultrasound (EUS) should play important roles in detecting abnormal image findings, such as local irregular MPD stenosis, caliber MPD changes, small cystic lesions, or branch duct dilatation. Additionally, EUS could detect hypoechoic areas around the MPD stenosis in some patients with HG-PanIN. Subsequently, endoscopic retrograde cholangiopancreatography (ERCP) and its associated pancreatic juice cytology, including serial pancreatic juice aspiration cytologic examination (SPACE) after placement of an endoscopic nasopancreatic drainage (ENPD) tube, may have high diagnostic accuracy for confirming the malignancy in HG-PanIN. Although ERCP and its associated pancreatic cytology, including SPACE, may be associated with post-ERCP pancreatitis (PEP), a recent randomized trial suggested that a 4-Fr ENPD tube may reduce the incidence of PEP. In the future, further prospective multicenter studies are required to establish a standard method of SPACE. Additionally, further studies for novel biomarkers could help to establish evolutionary methods with duodenal fluid and pancreatic juice for the early and accurate diagnosis of early-stage PDAC.
Collapse
Affiliation(s)
- Keiji Hanada
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| | - Akihiro Shimizu
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| | - Keisuke Kurihara
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| | - Morito Ikeda
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| | - Takuya Yamamoto
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| | - Yasuhiro Okuda
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| | - Susumu Tazuma
- Department of Gastroenterology, Onomichi General Hospital, Hiroshima, Japan
| |
Collapse
|
9
|
Choi MH, Tjora E, Forthun RB, Engjom T, Ræder H, Hovland R, Molven A. KRAS mutation analysis by droplet digital PCR of duodenal juice from patients with MODY8 and other pancreatic diseases. Pancreatology 2021; 21:1460-1465. [PMID: 34580018 DOI: 10.1016/j.pan.2021.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/19/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Maturity-onset diabetes of the young type 8 (MODY8 or CEL-MODY) is an inherited pancreatic disease characterized by chronic inflammation of the pancreas and diabetes. It is not known whether MODY8 patients have increased risk for developing pancreatic cancer. We investigated KRAS mutation load in duodenal juice from MODY8 patients, comparing with other groups of pancreatic disease. METHODS Droplet digital PCR (ddPCR) was used to detect KRAS codon 12/13/61 mutations in duodenal juice sampled from 11 MODY8 patients, nine healthy subjects and 100 patients clinically investigated due to suspected pancreatic disease. RESULTS KRAS mutations were detected in 4/11 patients with MODY8 (36%), 1/9 healthy subjects (11%), 15/44 patients with chronic pancreatitis (CP, 34%), 3/5 patients with pancreatic ductal adenocarcinoma (PDAC, 60%), 3/20 patients with acute pancreatitis (15%), 0/13 patients with other pancreatic disorders and 2/18 patients with nonpancreatic gastrointestinal disease (11%). Of the 28 positive juice samples, 25 (89%) had low-abundance mutations in codons 12/13, with a variant allele frequency (VAF) less than 1%. KRAS-positive patients with MODY8 or CP had significantly lower VAFs than patients with PDAC (Mann-Whitney U test; p = 0.041). Although the overall mutation detection rate was higher for subjects ≥50 years old (26%) than for younger subjects (15%), the difference was not statistically significant. CONCLUSIONS KRAS mutations were detectable in duodenal juice from MODY8 patients, but with low abundance and at the same frequency as in CP patients. The discriminative value of the analysis with regard to other pancreatic disease was limited.
Collapse
Affiliation(s)
- Man Hung Choi
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Erling Tjora
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway; Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Trond Engjom
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Helge Ræder
- Department of Pediatrics, Haukeland University Hospital, Bergen, Norway; Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Randi Hovland
- Section for Tumor Genomics, Haukeland University Hospital, Bergen, Norway; Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
10
|
Yousaf MN, Chaudhary FS, Ehsan A, Suarez AL, Muniraj T, Jamidar P, Aslanian HR, Farrell JJ. Endoscopic ultrasound (EUS) and the management of pancreatic cancer. BMJ Open Gastroenterol 2021; 7:bmjgast-2020-000408. [PMID: 32414753 PMCID: PMC7232396 DOI: 10.1136/bmjgast-2020-000408] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer-related mortality in western countries. Early diagnosis of pancreatic cancers plays a key role in the management by identification of patients who are surgical candidates. The advancement in the radiological imaging and interventional endoscopy (including endoscopic ultrasound (EUS), endoscopic retrograde cholangiopancreatography and endoscopic enteral stenting techniques) has a significant impact in the diagnostic evaluation, staging and treatment of pancreatic cancer. The multidisciplinary involvement of radiology, gastroenterology, medical oncology and surgical oncology is central to the management of patients with pancreatic cancers. This review aims to highlight the diagnostic and therapeutic role of EUS in the management of patients with pancreatic malignancy, especially pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Muhammad Nadeem Yousaf
- Department of Medicine, Section Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA .,Department of Internal Medicine, MedStar Union Memorial Hospital, Baltimore, Maryland, USA.,Department of Internal Medicine, MedStar Good Samaritan Hospital, Baltimore, Maryland, USA.,Department of Internal Medicine, MedStar Franklin Square Medical Center, Baltimore, Maryland, USA
| | - Fizah S Chaudhary
- Department of Internal Medicine, MedStar Union Memorial Hospital, Baltimore, Maryland, USA.,Department of Internal Medicine, MedStar Good Samaritan Hospital, Baltimore, Maryland, USA.,Department of Internal Medicine, MedStar Franklin Square Medical Center, Baltimore, Maryland, USA
| | - Amrat Ehsan
- Department of Internal Medicine, MedStar Union Memorial Hospital, Baltimore, Maryland, USA.,Department of Internal Medicine, MedStar Good Samaritan Hospital, Baltimore, Maryland, USA.,Department of Internal Medicine, MedStar Franklin Square Medical Center, Baltimore, Maryland, USA
| | - Alejandro L Suarez
- Department of Medicine, Section Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thiruvengadam Muniraj
- Department of Medicine, Section Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Priya Jamidar
- Department of Medicine, Section Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Harry R Aslanian
- Department of Medicine, Section Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - James J Farrell
- Department of Medicine, Section Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
11
|
Mohanty A, Mohanty SK, Rout S, Pani C. Liquid Biopsy, the hype vs. hope in molecular and clinical oncology. Semin Oncol 2021; 48:259-267. [PMID: 34384614 DOI: 10.1053/j.seminoncol.2021.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 05/28/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022]
Abstract
The molecular landscape of tumors has been traditionally established using a biopsy or resection specimens. These modalities result in sampling bias that offer only a single snapshot of tumor heterogeneity. Over the last decade intensive research towards alleviating such a bias and obtaining an integral yet accurate portrait of the tumors, evolved to the use of established molecular and genetic analysis using blood and several other body fluids, such as urine, saliva, and pleural effusions as liquid biopsies. Genomic profiling of the circulating markers including circulating cell-free tumor DNA (ctDNA), circulating tumor cells (CTCs) or even RNA, proteins, and lipids constituting exosomes, have facilitated the diligent monitoring of response to treatment, allowed one to follow the emergence of drug resistance, and enumerate minimal residual disease. The prevalence of tumor educated platelets (TEPs) and our understanding of how tumor cells influence platelets are beginning to unearth TEPs as a potentially dynamic component of liquid biopsies. Here, we review the biology, methodology, approaches, and clinical applications of biomarkers used to assess liquid biopsies. The current review addresses recent technological advances and different forms of liquid biopsy along with upcoming challenges and how they can be integrated to get the best possible tumor-derived genetic information that can be leveraged to more precise therapies for patient as liquid biopsies become increasingly routine in clinical practice.
Collapse
Affiliation(s)
- Abhishek Mohanty
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India.
| | - Sambit K Mohanty
- Advanced Medical Research Institute, Bhubaneswar, Odisha, India; CORE Diagnostics, Gurgaon, Haryana, India
| | - Sipra Rout
- Christian Medical College, Vellore, Tamil Nadu, India
| | | |
Collapse
|
12
|
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
|
13
|
Sagami R, Yamao K, Nakahodo J, Minami R, Tsurusaki M, Murakami K, Amano Y. Pre-Operative Imaging and Pathological Diagnosis of Localized High-Grade Pancreatic Intra-Epithelial Neoplasia without Invasive Carcinoma. Cancers (Basel) 2021; 13:cancers13050945. [PMID: 33668239 PMCID: PMC7956417 DOI: 10.3390/cancers13050945] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/08/2021] [Accepted: 02/19/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) arises from precursor lesions, such as pancreatic intra-epithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN). The prognosis of high-grade precancerous lesions, including high-grade PanIN and high-grade IPMN, without invasive carcinoma is good, despite the overall poor prognosis of PDAC. High-grade PanIN, as a lesion preceding invasive PDAC, is therefore a primary target for intervention. However, detection of localized high-grade PanIN is difficult when using standard radiological approaches. Therefore, most studies of high-grade PanIN have been conducted using specimens that harbor invasive PDAC. Recently, imaging characteristics of high-grade PanIN have been revealed. Obstruction of the pancreatic duct due to high-grade PanIN may induce a loss of acinar cells replaced by fibrosis and lobular parenchymal atrophy. These changes and additional inflammation around the branch pancreatic ducts (BPDs) result in main pancreatic duct (MPD) stenosis, dilation, retention cysts (BPD dilation), focal pancreatic parenchymal atrophy, and/or hypoechoic changes around the MPD. These indirect imaging findings have become important clues for localized, high-grade PanIN detection. To obtain pre-operative histopathological confirmation of suspected cases, serial pancreatic-juice aspiration cytologic examination is effective. In this review, we outline current knowledge on imaging characteristics of high-grade PanIN.
Collapse
Affiliation(s)
- Ryota Sagami
- Department of Gastroenterology, Oita San-ai Medical Center, 1213 Oaza Ichi, Oita, Oita 870-1151, Japan
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Correspondence: ; Tel.: +81-97-541-1311; Fax: +81-97-541-5218
| | - Kentaro Yamao
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Department of Gastroenterology and Hepatology, Kindai University, Osaka-Sayama, Osaka 589-8511, Japan
| | - Jun Nakahodo
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Department of Gastroenterology Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo 113-8677, Japan
| | - Ryuki Minami
- Pancreatic Cancer Research for Secure Salvage Young Investigators (PASSYON), Osaka-Sayama, Osaka 589-8511, Japan; (K.Y.); (J.N.); (R.M.)
- Department of Gastroenterology, Tenri Hospital, 200 Mishimacho, Tenri, Nara 632-0015, Japan
| | - Masakatsu Tsurusaki
- Department of Diagnostic Radiology, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka 589-8511, Japan;
| | - Kazunari Murakami
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasamacho, Yufu, Oita 879-5593, Japan;
| | - Yuji Amano
- Department of Endoscopy, Urawa Kyosai Hospital, 3-15-31 Harayama, Midoriku, Saitama 336-0931, Japan;
| |
Collapse
|
14
|
Ideno N, Mori Y, Nakamura M, Ohtsuka T. Early Detection of Pancreatic Cancer: Role of Biomarkers in Pancreatic Fluid Samples. Diagnostics (Basel) 2020; 10:diagnostics10121056. [PMID: 33291257 PMCID: PMC7762187 DOI: 10.3390/diagnostics10121056] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/28/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related deaths worldwide. Most patients with PDAC present with symptomatic, surgically unresectable disease. Therefore, the establishment of strategies for the early detection is urgently needed. Molecular biomarkers might be useful in various phases of a strategy to identify high-risk individuals in the general population and to detect high-risk lesions during intense surveillance programs combined with imaging modalities. However, the low sensitivity and specificity of biomarkers currently available for PDAC, such as carbohydrate 19-9 (CA19-9), contribute to the late diagnosis of this deadly disease. Although almost all classes of biomarker assays have been studied, most of them are used in the context of symptomatic diseases. Compared to other body fluids, pancreatic juice and duodenal fluid are better sources of DNA, RNA, proteins, and exosomes derived from neoplastic cells and have the potential to increase the sensitivity/specificity of these biomarkers. The number of studies using duodenal fluid with or without secretin stimulation for DNA/protein marker tests have been increasing because of the less-invasiveness in comparison to pancreatic juice collection by endoscopic retrograde cholangiopancreatography (ERCP) and endoscopic ultrasound-guided fine needle aspiration (EUS-FNA). Genomic analyses have been very well-studied, and based on PDAC progression model, mutations detected in pancreatic juice/duodenal fluid seem to indicate the presence of microscopic precursors and high-grade dysplasia/invasive cancer. In addition to known proteins overexpressed both in precursors and PDACs, such as CEA and S100P, comprehensive proteomic analysis of pancreatic juice from patients with PDAC identified many proteins which were not previously described. A novel technique to isolate exosomes from pancreatic juice was recently invented and identification of exosomal microRNA’s 21 and 155 could be biomarkers for diagnosis of PDAC. Since many studies have explored biomarkers in fluid samples containing pancreatic juice and reported excellent diagnostic accuracy, we need to discuss how these biomarker assays can be validated and utilized in the strategy of early detection of PDAC.
Collapse
Affiliation(s)
- Noboru Ideno
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (N.I.); (Y.M.); (M.N.)
| | - Yasuhisa Mori
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (N.I.); (Y.M.); (M.N.)
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (N.I.); (Y.M.); (M.N.)
| | - Takao Ohtsuka
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical Sciences, Kagoshima University, Kagoshima 890-8520, Japan
- Correspondence: ; Tel.: +81-99-275-5361
| |
Collapse
|
15
|
The Diagnostic Accuracy of Mutant KRAS Detection from Pancreatic Secretions for the Diagnosis of Pancreatic Cancer: A Meta-Analysis. Cancers (Basel) 2020; 12:cancers12092353. [PMID: 32825312 PMCID: PMC7564395 DOI: 10.3390/cancers12092353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
This meta-analysis aims to identify the diagnostic accuracy of mutations in the Kirsten Rat Sarcoma (KRAS) oncogene in the diagnosis of pancreatic ductal adenocarcinoma (PDAC). The survival of PDAC remains poor often due to the fact that disease is advanced at diagnosis. We analysed 22 studies, with a total of 2156 patients, to identify if the detection of KRAS mutations from pancreatic exocrine secretions yields sufficient specificity and sensitivity to detect patients with PDAC amongst healthy individuals. The majority of the studies were retrospective, samples were obtained endoscopically or surgically, and included comparator populations of patients with chronic pancreatitis and pre-malignant pancreatic lesions (PanIN) as well as healthy controls. We performed several analyses to identify the diagnostic accuracy for PDAC among these patient populations. Our results highlighted that the diagnostic accuracy of KRAS mutation for PDAC was of variable sensitivity and specificity when compared with PanINs and chronic pancreatitis, but had a higher specificity among healthy individuals. The sensitivity of this test must be improved to prevent missing early PDAC or PanINs. This could be achieved with rigorous prospective cohort studies, in which high-risk patients with normal cross-sectional imaging undergo surveillance following KRAS mutation testing.
Collapse
|
16
|
de la Fuente J, Majumder S. Molecular Diagnostics and Testing for Pancreatic Cysts. CURRENT TREATMENT OPTIONS IN GASTROENTEROLOGY 2020; 18:158-171. [PMID: 31989386 DOI: 10.1007/s11938-020-00270-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW In current clinical practice, the diagnosis and management of pancreatic cystic lesions (PCLs) are based on guidelines that combine clinical and imaging findings. These guidelines usefully identify a large category of low-risk PCLs that do not require treatment. However, they have limited accuracy for diagnosis of advanced neoplasia in worrisome and high-risk PCLs. Novel molecular markers that can accurately detect advanced neoplasia in PCLs can transform the care of patients with PCLs. We reviewed the recent medical literature on molecular diagnostics of PCLs and summarized molecular biomarkers assayed in cyst fluid, pancreatic juice, and blood. RECENT FINDINGS Several studies have been recently published describing promising early results in genetic, epigenetic, and protein biomarkers from cyst fluid to help in both histologic diagnosis and detection of advanced neoplasia. The majority of studies have been completed using opportunistically collected archival cyst fluid and few report validation in independent sample sets. Results of ongoing multicenter prospective validation studies are awaited and will help define the best combination of cyst fluid molecular markers. In multifocal PCLs communicating with the pancreatic ductal system, a pancreatic juice biomarker is likely to be less invasive and more informative. Novel biomarkers in pancreatic juice and blood are in early phases of study. SUMMARY The field of molecular diagnostic biomarkers for PCLs is rapidly evolving with several promising candidate markers being prospectively evaluated. In the near future, these novel molecular markers, combined with advances in imaging technology, will transform clinical decision-making in the management of PCLs and improve patient outcomes.
Collapse
Affiliation(s)
- Jaime de la Fuente
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Sciences, 200 First St SW, Rochester, MN, 55905, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Sciences, 200 First St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
17
|
Llach J, Carballal S, Moreira L. Familial Pancreatic Cancer: Current Perspectives. Cancer Manag Res 2020; 12:743-758. [PMID: 32099470 PMCID: PMC6999545 DOI: 10.2147/cmar.s172421] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/15/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer (PC) is a highly lethal disease, mostly incurable when detected. Thus, despite advances in PC treatments, only around 7% of patients survive 5-years after diagnosis. This morbid outcome is secondary to multifactorial reasons, such as late-stage diagnosis, rapid progression and minimal response to chemotherapy. Based on these factors, it is of special relevance to identify PC high-risk individuals in order to establish preventive and early detection measures. Although most PC are sporadic, approximately 10% cases have a familial basis. No main causative gene of PC has been identified but several known germline pathogenic mutations are related with an increased risk of this tumor. These inherited cancer syndromes represent 3% of all PC. On the other hand, in 7% of cases of PC, there is a strong family history without a causative germline mutation, a situation known as familial pancreatic cancer (FPC). In recent years, there is increasing evidence supporting the benefit of genetic germline analysis in PC patients, and periodic pancreatic screening in PC high-risk patients (mainly those with a lifetime risk greater than 5%), although there is no general agreement in the group of patients and individuals to study and screen. In the present review, we expose an update in the field of hereditary and FPC, with the aim of describing the current strategies and implications in genetic counseling, surveillance and therapeutic interventions.
Collapse
Affiliation(s)
- Joan Llach
- Departmento de Gastroenterología, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d' Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| | - Sabela Carballal
- Departmento de Gastroenterología, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d' Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| | - Leticia Moreira
- Departmento de Gastroenterología, Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d' Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Universidad de Barcelona, Barcelona, Spain
| |
Collapse
|
18
|
Zhou H, Zhu Y, Wei F, Shao Y, Pan J, Wang G, Xu K, Cheng Y. Significance of MUC2 gene methylation detection in pancreatic cancer diagnosis. Pancreatology 2019; 19:1049-1053. [PMID: 31590960 DOI: 10.1016/j.pan.2019.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 09/15/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE This study was conducted to explore the diagnostic value of MUC2 gene methylation in pancreatic cancer. METHODS Methylation restriction enzyme digestion (Msp I/Hap II) and polymerase chain reaction (PCR) were performed to detect methylation of the MUC2 gene in fecal and blood specimens from seven study subjects with pancreatic cancer (PC), chronic pancreatitis (CP), or normal controls (CON). Simultaneously, blood CA 19-9 levels were detected as a positive indicator of PC. RESULTS MUC2 methylation was detected in 50% of PC cell lines. In fecal samples, the MUC2 methylation rate in PC (n = 30) was 43.3%, which was significantly higher than those in CP (n = 8, 0%, P < 0.05) and CON (n = 20, 5.0%, P < 0.05). In blood samples, the MUC2 methylation rate in PC (n = 40) was 52.5%, which was significantly higher than those in CP (n = 15, 0%, P < 0.01) and CON (n = 25, 4.0%, P < 0.01). For PC diagnosis, MUC2 gene methylation in blood samples showed higher specificity and positive predictive value than CA 19-9. The combined detection in the feces and blood showed a 60% MUC2 methylation rate in PC (n = 10), which was higher than those in the CP (n = 5, 0%, P < 0.01) and CON (n = 12, 0%, P < 0.01). CONCLUSIONS The study can clearly indicate that combined detection of MUC2 gene methylation in the peripheral blood and feces could be used as a new screening and early diagnosis method for pancreatic cancer.
Collapse
Affiliation(s)
- Hanyu Zhou
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yingwei Zhu
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Feifei Wei
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yueting Shao
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Jinjin Pan
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Ge Wang
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Kequn Xu
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China.
| | - Yuqing Cheng
- Department of Oncology, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China.
| |
Collapse
|
19
|
Mateos RN, Nakagawa H, Hirono S, Takano S, Fukasawa M, Yanagisawa A, Yasukawa S, Maejima K, Oku-Sasaki A, Nakano K, Dutta M, Tanaka H, Miyano S, Enomoto N, Yamaue H, Nakai K, Fujita M. Genomic analysis of pancreatic juice DNA assesses malignant risk of intraductal papillary mucinous neoplasm of pancreas. Cancer Med 2019; 8:4565-4573. [PMID: 31225717 PMCID: PMC6712468 DOI: 10.1002/cam4.2340] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 12/14/2022] Open
Abstract
Intraductal papillary mucinous neoplasm (IPMN) of pancreas has a high risk to develop into invasive cancer or co‐occur with malignant lesion. Therefore, it is important to assess its malignant risk by less‐invasive approach. Pancreatic juice cell‐free DNA (PJD) would be an ideal material in this purpose, but genetic biomarkers for predicting malignant risk from PJD are not yet established. We here performed deep exome sequencing analysis of PJD from 39 IPMN patients with or without malignant lesion. Somatic alterations and copy number alterations (CNAs) detected in PJD were compared with the histologic grade of IPMN to evaluate their potential as a malignancy marker. Somatic mutations of KRAS, GNAS, TP53, and RNF43 were commonly detected in PJD of IPMNs, but no association with the histologic grades of IPMN was found. Instead, mutation burden was positively correlated with the histologic grade (r = 0.427, P = 0.015). We also observed frequent copy number deletions in 17p13 (TP53) and amplifications in 7q21 and 8q24 (MYC) in PJDs. The amplifications in 7q21 and 8q24 were positively correlated with the histologic grade and most prevalent in the cases of invasive carcinoma (P = 0.002 and 7/11; P = 0.011 and 6/11, respectively). We concluded that mutation burden and CNAs detected in PJD may have potential to assess the malignant progression risk of IPMNs.
Collapse
Affiliation(s)
- Raúl N Mateos
- Department of Computational Biology and Medical Science, The University of Tokyo, Chiba, Japan.,Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Seiko Hirono
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Takano
- First Department of Internal Medicine, University of Yamanashi, Yamanashi, Japan
| | - Mitsuharu Fukasawa
- First Department of Internal Medicine, University of Yamanashi, Yamanashi, Japan
| | - Akio Yanagisawa
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoru Yasukawa
- Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Aya Oku-Sasaki
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kaoru Nakano
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Munmee Dutta
- Department of Computational Biology and Medical Science, The University of Tokyo, Chiba, Japan.,Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroko Tanaka
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- Laboratory of DNA Information Analysis, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Nobuyuki Enomoto
- First Department of Internal Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Science, The University of Tokyo, Chiba, Japan.,Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masashi Fujita
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
20
|
Pratt ED, Cowan RW, Manning SL, Qiao E, Cameron H, Schradle K, Simeone DM, Zhen DB. Multiplex Enrichment and Detection of Rare KRAS Mutations in Liquid Biopsy Samples using Digital Droplet Pre-Amplification. Anal Chem 2019; 91:7516-7523. [DOI: 10.1021/acs.analchem.8b01605] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Erica D. Pratt
- Ahmed Center for Pancreatic Cancer Research, Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Robert W. Cowan
- Ahmed Center for Pancreatic Cancer Research, Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Sara L. Manning
- Ahmed Center for Pancreatic Cancer Research, Department of Gastroenterology, Hepatology and Nutrition, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | | | | | | | | |
Collapse
|
21
|
Wood LD, Yurgelun MB, Goggins MG. Genetics of Familial and Sporadic Pancreatic Cancer. Gastroenterology 2019; 156:2041-2055. [PMID: 30660730 DOI: 10.1053/j.gastro.2018.12.039] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/14/2018] [Accepted: 12/05/2018] [Indexed: 12/14/2022]
Abstract
In the previous decade, comprehensive genomic analyses have yielded important insights about the genetic alterations that underlie pancreatic tumorigenesis. Whole-exome and whole-genome sequencing of pancreatic ductal adenocarcinomas have confirmed the critical driver genes altered in the majority of pancreatic cancers, as well as identified numerous less frequently altered driver genes, and have delineated cancer subgroups with unique biological and clinical features. It is now appreciated that pancreatic susceptibility gene alterations are often identified in patients with pancreatic cancer without family histories suggestive of a familial cancer syndrome, prompting recent efforts to expand gene testing to all patients with pancreatic cancer. Studies of pancreatic cancer precursor lesions have begun to elucidate the evolutionary history of pancreatic tumorigenesis and to help us understand the utility of biomarkers for early detection and targets to develop new therapeutic strategies. In this review, we discuss the results of comprehensive genomic characterization of pancreatic ductal adenocarcinoma and its precursor lesions, and we highlight translational applications in early detection and therapy.
Collapse
Affiliation(s)
- Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts; Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Michael G Goggins
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Medicine, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
22
|
Reza J, Almodovar AJ, Srivastava M, Veldhuis PP, Patel S, Fanaian N, Zhu X, Litherland SA, Arnoletti JP. K-RAS Mutant Gene Found in Pancreatic Juice Activated Chromatin From Peri-ampullary Adenocarcinomas. Epigenet Insights 2019; 12:2516865719828348. [PMID: 30815628 PMCID: PMC6383091 DOI: 10.1177/2516865719828348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
External pancreatic duct stents inserted after resection of pancreatic head
tumors provide unique access to pancreatic juice analysis of genetic and
metabolic components that may be associated with peri-ampullary tumor
progression. For this pilot study, portal venous blood and pancreatic juice
samples were collected from 17 patients who underwent pancreaticoduodenectomy
for peri-ampullary tumors. Portal vein circulating tumor cells (CTC) were
isolated by high-speed fluorescence-activated cell sorting (FACS) and analyzed
by quantitative reverse transcription polymerase chain reaction (RT-PCR) for
K-RAS exon 12 mutant gene expression
(K-RASmut). DNA, chromatin, and histone acetylated active
chromatin were isolated from pancreatic juice samples by chromatin
immunoprecipitation (ChIP) and the presence of K-RASmut and
other cancer-related gene sequences detected by quantitative polymerase chain
reaction (PCR) and ChIP-Seq. Mutated K-RAS gene was detectable
in activated chromatin in pancreatic juice secreted after surgical resection of
pancreatic, ampullary and bile duct carcinomas and directly correlated with the
number of CTC found in the portal venous blood (P = .0453).
ChIP and ChIP-Seq detected acetylated chromatin in peri-ampullary cancer patient
juice containing candidate chromatin loci, including RET
proto-oncogene, not found in similar analysis of pancreatic juice from
non-malignant ampullary adenoma. The presence of active tumor cell chromatin in
pancreatic juice after surgical removal of the primary tumor suggests that
viable cancer cells either remain or re-emerge from the remnant pancreatic duct,
providing a potential source for tumor recurrence and cancer relapse. Therefore,
epigenetic analysis for active chromatin in pancreatic juice and portal venous
blood CTC may be useful for prognostic risk stratification and potential
identification of molecular targets in peri-ampullary cancers.
Collapse
Affiliation(s)
- Joseph Reza
- General Surgery Residency Program, AdventHealth, Orlando, FL, USA
| | - Alvin Jo Almodovar
- Translational Research, Cancer Institute, AdventHealth, Orlando, FL, USA
| | - Milan Srivastava
- Translational Research, Cancer Institute, AdventHealth, Orlando, FL, USA
| | - Paula P Veldhuis
- Institute for Surgical Advancement, AdventHealth, Orlando, FL, USA
| | - Swati Patel
- Institute for Surgical Advancement, AdventHealth, Orlando, FL, USA
| | - Na'im Fanaian
- Center for Diagnostic Pathology, AdventHealth, Orlando, FL, USA
| | - Xiang Zhu
- Center for Interventional Endoscopy, AdventHealth, Orlando, FL, USA
| | - Sally A Litherland
- Translational Research, Cancer Institute, AdventHealth, Orlando, FL, USA
| | - J Pablo Arnoletti
- Translational Research, Cancer Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
23
|
de Mestier L, Muller M, Cros J, Vullierme MP, Vernerey D, Maire F, Dokmak S, Rebours V, Sauvanet A, Lévy P, Hammel P. Appropriateness of pancreatic resection in high-risk individuals for familial pancreatic ductal adenocarcinoma: a patient-level meta-analysis and proposition of the Beaujon score. United European Gastroenterol J 2019; 7:358-368. [PMID: 31019704 DOI: 10.1177/2050640618824910] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
Background About 5% of pancreatic ductal adenocarcinomas are inherited due to a deleterious germline mutation detected in 20% or fewer families. Pancreatic screening in high-risk individuals is proposed to allow early surgical treatment of (pre)malignant lesions. The outcomes of pancreatic surgery in high-risk individuals have never been correctly explored. Objectives To evaluate surgical appropriateness and search for associated factors in high-risk individuals. Methods A patient-level meta-analysis was performed including studies published since 1999. Individual classification distinguished the highest risk imaging abnormality into low-risk or high-risk abnormality, and the highest pathological degree of malignancy of lesions into no/low malignant potential or potentially/frankly malignant. Surgical appropriateness was considered when potentially/frankly malignant lesions were resected. Results Thirteen out of 24 studies were selected, which reported 90 high-risk individuals operated on. Low-risk/high-risk abnormalities were preoperatively detected in 46.7%/53.3% of operated high-risk individuals, respectively. Surgical appropriateness was consistent in 38 (42.2%) high-risk individuals, including 20 pancreatic ductal adenocarcinomas (22.2%). Identification of high-risk abnormalities was strongly associated with surgical appropriateness at multivariate analysis (P = 0.001). We proposed a score and nomogram predictive of surgical appropriateness, including high-risk abnormalities, age and existence of deleterious germline mutation. Conclusion Overall, 42.2% of high-risk individuals underwent appropriate surgery. The proposed score might help selecting the best candidates among high-risk individuals for pancreatic resection.
Collapse
Affiliation(s)
- Louis de Mestier
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Marie Muller
- Department of Digestive Oncology and Genetic Counselling, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Jérôme Cros
- Department of Pathology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Marie-Pierre Vullierme
- Department of Radiology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Dewi Vernerey
- Department of Methodology and Quality of Life in Oncology Unit, EA 3181, Minjoz University Hospital, Besançon, France
| | - Frédérique Maire
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Safi Dokmak
- Department of Hepatobiliary and Pancreatic Surgery, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Vinciane Rebours
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Alain Sauvanet
- Department of Hepatobiliary and Pancreatic Surgery, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Philippe Lévy
- Department of Gastroenterology and Pancreatology, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| | - Pascal Hammel
- Department of Digestive Oncology and Genetic Counselling, Beaujon Hospital (AP-HP) and University Paris Diderot, Clichy, France
| |
Collapse
|
24
|
Choi MH, Mejlænder-Andersen E, Manueldas S, El Jellas K, Steine SJ, Tjensvoll K, Sætran HA, Knappskog S, Hoem D, Nordgård O, Hovland R, Molven A. Mutation analysis by deep sequencing of pancreatic juice from patients with pancreatic ductal adenocarcinoma. BMC Cancer 2019; 19:11. [PMID: 30611220 PMCID: PMC6321709 DOI: 10.1186/s12885-018-5195-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
Background Reliable methods are needed to identify patients with early-stage cancer or high-grade precancerous lesions in the pancreas. Analysis of pancreatic juice to detect somatic mutations could represent one such approach. Here we investigated the concordance between mutations found in the primary tumor and pancreatic juice from the same patient. Methods Amplicon-based targeted deep sequencing was performed on samples from 21 patients with pancreatic ductal adenocarcinoma (PDAC) who had undergone Whipple’s operation. Mutation profiles were determined in formalin-fixed sections of the primary tumor and in pancreatic juice sampled from the main pancreatic duct during surgery. Results Using a cut-off of 3% for variant allele frequency, KRAS mutations were detected in 20/21 primary tumors (95%) and in 15/21 (71%) juice samples. When also considering low-frequency variants, KRAS mutations were found in 20/21 juice samples. Most juice samples exhibited multiple KRAS variants not seen in the primary tumor, and only in 11 cases (52%) did the most abundant variant of the juice correspond to the KRAS mutation detected in the tumor. TP53 mutations were found in 16 tumors (76%) and six juice samples (29%). Among the positive juice samples, only one exhibited more than a single TP53 mutation. Detection of both KRAS and TP53 mutations was fully concordant in the primary tumor and juice sample in 7/21 cases (33%). Conclusions Pancreatic juice from PDAC patients is rich in KRAS mutations often not seen in the primary tumor and possibly reflecting precancerous lesions in other regions of the pancreas. The inclusion of TP53 mutation detection and additional markers must therefore be considered for fully exploiting the clinical potential of pancreatic juice samples in early cancer detection. Electronic supplementary material The online version of this article (10.1186/s12885-018-5195-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Man Hung Choi
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Eline Mejlænder-Andersen
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Sophia Manueldas
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Khadija El Jellas
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Solrun J Steine
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kjersti Tjensvoll
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Hege Aase Sætran
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Stian Knappskog
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Dag Hoem
- Department of Gastrointestinal Surgery, Haukeland University Hospital, Bergen, Norway
| | - Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Randi Hovland
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Anders Molven
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway. .,Department of Pathology, Haukeland University Hospital, Bergen, Norway. .,KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
25
|
Quan S, Principe DR, Dean AE, Park SH, Grippo PJ, Gius D, Horikoshi N. Loss of Sirt2 increases and prolongs a caerulein-induced pancreatitis permissive phenotype and induces spontaneous oncogenic Kras mutations in mice. Sci Rep 2018; 8:16501. [PMID: 30405152 PMCID: PMC6220268 DOI: 10.1038/s41598-018-34792-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Mice lacking Sirt2 spontaneously develop tumors in multiple organs, as well as when expressed in combination with oncogenic KrasG12D, leading to pancreatic tumors. Here, we report that after caerulein-induced pancreatitis, Sirt2-deficient mice exhibited an increased inflammatory phenotype and delayed pancreatic tissue recovery. Seven days post injury, the pancreas of Sirt2-/- mice display active inflammation, whereas wild-type mice had mostly recovered. In addition, the pancreas from the Sirt2-/- mice exhibited extensive tissue fibrosis, which was still present at six weeks after exposure. The mice lacking Sirt2 also demonstrated an enhanced whole body pro-inflammatory phenotype that was most obvious with increasing age. Importantly, an accumulation of a cell population with spontaneous cancerous KrasG12D mutations was observed in the Sirt2-/- mice that is enhanced in the recovering pancreas after exposure to caerulein. Finally, transcriptome analysis of the pancreas of the Sirt2-/- mice exhibited a pro-inflammatory genomic signature. These results suggest that loss of Sirt2, as well as increased age, enhanced the immune response to pancreatic injury and induced an inflammatory phenotype permissive for the accumulation of cells carrying oncogenic Kras mutations.
Collapse
Affiliation(s)
- Songhua Quan
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daniel R Principe
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Angela E Dean
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Seong-Hoon Park
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of General and Applied Toxicology, Genetic Toxicology Research Group, Korea Institute of Toxicology (KIT), Daejeon, South Korea
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - David Gius
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Pharmacology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Bartsch DK, Gercke N, Strauch K, Wieboldt R, Matthäi E, Wagner V, Rospleszcz S, Schäfer A, Franke FS, Mintziras I, Bauer C, Grote T, Figiel J, Di Fazio P, Burchert A, Reinartz S, Pogge von Strandmann E, Klöppel G, Slater EP. The Combination of MiRNA-196b, LCN2, and TIMP1 is a Potential Set of Circulating Biomarkers for Screening Individuals at Risk for Familial Pancreatic Cancer. J Clin Med 2018; 7:295. [PMID: 30241369 PMCID: PMC6210952 DOI: 10.3390/jcm7100295] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
Individuals at risk (IAR) of familial pancreatic cancer (FPC) are good candidates for screening. Unfortunately, neither reliable imaging modalities nor biomarkers are available to detect high-grade precursor lesions or early cancer. Circulating levels of candidate biomarkers LCN2, TIMP1, Glypican-1, RNU2-1f, and miRNA-196b were analyzed in 218 individuals with sporadic pancreatic ductal adenocarcinoma (PDAC, n = 50), FPC (n = 20), chronic pancreatitis (n = 10), IAR with relevant precursor lesions (n = 11) or non-relevant lesions (n = 5), 20 controls, and IAR with (n = 51) or without (n = 51) lesions on pancreatic imaging. In addition, corresponding duodenal juice samples were analyzed for Glypican-1 (n = 144) enrichment and KRAS mutations (n = 123). The panel miR-196b/LCN2/TIMP1 could distinguish high-grade lesions and stage I PDAC from controls with absolute specificity and sensitivity. In contrast, Glypican-1 enrichment in serum exosomes and duodenal juice was not diagnostic. KRAS mutations in duodenal juice were detected in 9 of 12 patients with PDAC and only 4 of 9 IAR with relevant precursor lesions. IAR with lesions on imaging had elevated miR-196b/LCN2/TIMP1 levels (p = 0.0007) and KRAS mutations in duodenal juice (p = 0.0004) significantly more often than IAR without imaging lesions. The combination miR-196b/LCN2/TIMP1 might be a promising biomarker set for the detection of high-grade PDAC precursor lesions in IAR of FPC families.
Collapse
Affiliation(s)
- Detlef K Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Norman Gercke
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Konstantin Strauch
- Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Faculty of Medicine, Ludwig-Maximilians-Universität, Marchioninistr. 15, D-81377 Munich, Germany.
- Institute of Genetic Epidemiology, Helmholtz Zentrum München⁻German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany.
| | - Ronja Wieboldt
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Elvira Matthäi
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Vinona Wagner
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Susanne Rospleszcz
- Institut für Medizinische Informationsverarbeitung, Biometrie und Epidemiologie, Faculty of Medicine, Ludwig-Maximilians-Universität, Marchioninistr. 15, D-81377 Munich, Germany.
- Institute of Genetic Epidemiology, Helmholtz Zentrum München⁻German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany.
| | - Agnes Schäfer
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Frederike S Franke
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Ioannis Mintziras
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Christian Bauer
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Tobias Grote
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Jens Figiel
- Department of Diagnostic and Interventional Radiology, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| | - Silke Reinartz
- Center for Tumor and Immune Biology, Philipps University Marburg, Hans-Meerwein-Str. 3, D-35043 Marburg, Germany.
| | - Elke Pogge von Strandmann
- Center for Tumor and Immune Biology, Philipps University Marburg, Hans-Meerwein-Str. 3, D-35043 Marburg, Germany.
| | - Günter Klöppel
- Department of Pathology, Technical University Munich, Trogerstr. 18, D-81675 Munich, Germany.
| | - Emily P Slater
- Department of Visceral, Thoracic and Vascular Surgery, Philipps University Marburg, Baldingerstrasse, D-35043 Marburg, Germany.
| |
Collapse
|
27
|
Canto MI, Almario JA, Schulick RD, Yeo CJ, Klein A, Blackford A, Shin EJ, Sanyal A, Yenokyan G, Lennon AM, Kamel IR, Fishman EK, Wolfgang C, Weiss M, Hruban RH, Goggins M. Risk of Neoplastic Progression in Individuals at High Risk for Pancreatic Cancer Undergoing Long-term Surveillance. Gastroenterology 2018; 155:740-751.e2. [PMID: 29803839 PMCID: PMC6120797 DOI: 10.1053/j.gastro.2018.05.035] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 04/09/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Screening of individuals who have a high risk of pancreatic ductal adenocarcinoma (PDAC), because of genetic factors, frequently leads to identification of pancreatic lesions. We investigated the incidence of PDAC and risk factors for neoplastic progression in individuals at high risk for PDAC enrolled in a long-term screening study. METHODS We analyzed data from 354 individuals at high risk for PDAC (based on genetic factors of family history), enrolled in Cancer of the Pancreas Screening cohort studies at tertiary care academic centers from 1998 through 2014 (median follow-up time, 5.6 years). All subjects were evaluated at study entry (baseline) by endoscopic ultrasonography and underwent surveillance with endoscopic ultrasonography, magnetic resonance imaging, and/or computed tomography. The primary endpoint was the cumulative incidence of PDAC, pancreatic intraepithelial neoplasia grade 3, or intraductal papillary mucinous neoplasm with high-grade dysplasia (HGD) after baseline. We performed multivariate Cox regression and Kaplan-Meier analyses. RESULTS During the follow-up period, pancreatic lesions with worrisome features (solid mass, multiple cysts, cyst size > 3 cm, thickened/enhancing walls, mural nodule, dilated main pancreatic duct > 5 mm, or abrupt change in duct caliber) or rapid cyst growth (>4 mm/year) were detected in 68 patients (19%). Overall, 24 of 354 patients (7%) had neoplastic progression (14 PDACs and 10 HGDs) over a 16-year period; the rate of progression was 1.6%/year, and 93% had detectable lesions with worrisome features before diagnosis of the PDAC or HGD. Nine of the 10 PDACs detected during routine surveillance were resectable; a significantly higher proportion of patients with resectable PDACs survived 3 years (85%) compared with the 4 subjects with symptomatic, unresectable PDACs (25%), which developed outside surveillance (log rank P < .0001). Neoplastic progression occurred at a median age of 67 years; the median time from baseline screening until PDAC diagnosis was 4.8 years (interquartile range, 1.6-6.9 years). CONCLUSIONS In a long-term (16-year) follow-up study of individuals at high-risk for PDAC, we found most PDACs detected during surveillance (9/10) to be resectable, and 85% of these patients survived for 3 years. We identified radiologic features associated with neoplastic progression.
Collapse
Affiliation(s)
- Marcia Irene Canto
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland; Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland.
| | - Jose Alejandro Almario
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions,Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | | | | | - Alison Klein
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Amanda Blackford
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Eun Ji Shin
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Abanti Sanyal
- The Johns Hopkins Biostatistics Center, Johns Hopkins Bloomberg School of Public Health
| | - Gayane Yenokyan
- The Johns Hopkins Biostatistics Center, Johns Hopkins Bloomberg School of Public Health
| | - Anne Marie Lennon
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Ihab R. Kamel
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Elliot K. Fishman
- Department of Radiology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Christopher Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Matthew Weiss
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| | - Michael Goggins
- Department of Medicine (Gastroenterology), The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions,Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions
| |
Collapse
|
28
|
Arnoletti JP, Fanaian N, Reza J, Sause R, Almodovar AJ, Srivastava M, Patel S, Veldhuis PP, Griffith E, Shao YP, Zhu X, Litherland SA. Pancreatic and bile duct cancer circulating tumor cells (CTC) form immune-resistant multi-cell type clusters in the portal venous circulation. Cancer Biol Ther 2018; 19:887-897. [PMID: 30067440 DOI: 10.1080/15384047.2018.1480292] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Circulating tumor cells (CTC) enter the blood from many carcinomas and represent a likely source of metastatic dissemination. In contrast to the peripheral circulation, KRAS mutation- positive CTC thrive in the portal venous blood of patients with pancreatic ductal adenocarcinoma (PDAC). To analyze the essential interactions that contribute to carcinoma CTC growth and immune resistance, portal venous blood was collected during pancreatico-duodenectomy in 41 patients with peri-ampullary pathologies (PDAC = 11; ampullary adenocarcinoma (AA) = 15; distal cholangiocarcinoma (CC) = 6; IPMN = 7; non-malignant pancreatitis = 2). FACS-isolated cell populations from the portal circulation were reconstituted ex vivo using mixed cell reaction cultures (MCR). During the first 48hr, PDAC, AA, and CC patient CTC were all highly proliferative (mean 1.7 hr/cell cycle, 61.5% ± 20% growing cells) and resistant to apoptosis (mean 39% ± 25% apoptotic cells). PDAC CTC proliferation and resistance to T cell cytotoxicity were decreased among patients who received pre-operative chemotherapy (p = 0.0019, p = 0.0191, respectively). After 7 days in culture, CTC from PDAC, CC, and AA patients recruited multiple immune cell types, including CD105 + CD14 + myeloid fibroblasts, to organize into spheroid-like clusters. It was only in PDAC and CC-derived MCR that cluster formation promoted CTC survival, growth, and fibroblast differentiation. FACS depletion of CTC or myeloid fibroblast cells eliminated cluster network formation, and re-introduction of these cell populations reconstituted such ability. Our findings suggest that PDAC and CC CTC survival within the portal venous circulation is supported by their interactions with immune cells within multi-cell type clusters that could represent vectors of local recurrence and metastatic progression.
Collapse
Affiliation(s)
- J Pablo Arnoletti
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| | - Na'im Fanaian
- b Florida Hospital Diagnostic Pathology (NF) , Orlando , FL , USA
| | - Joseph Reza
- c General Surgery Residency Program , Orlando , FL , USA
| | - Ryan Sause
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| | - Alvin Jo Almodovar
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| | - Milan Srivastava
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| | - Swati Patel
- d Institute for Surgical Advancement , Orlando , FL , USA
| | | | - Elizabeth Griffith
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| | - Yai-Ping Shao
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| | - Xiang Zhu
- e Center for Interventional Endoscopy , Orlando , FL , USA
| | - Sally A Litherland
- a Departments of Translational Research (SAL, RS, AJOA, MS, EG, Y-PS); Surgical Oncology (JPA), Florida Hospital Cancer Institute , Orlando , FL , USA
| |
Collapse
|
29
|
Welinsky S, Lucas AL. Familial Pancreatic Cancer and the Future of Directed Screening. Gut Liver 2018; 11:761-770. [PMID: 28609837 PMCID: PMC5669591 DOI: 10.5009/gnl16414] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 11/01/2016] [Accepted: 11/01/2016] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer (PC) is the third most common cause of cancer-related death in the United States and the 12th most common worldwide. Mortality is high, largely due to late stage of presentation and suboptimal treatment regimens. Approximately 10% of PC cases have a familial basis. The major genetic defect has yet to be identified but may be inherited by an autosomal dominant pattern with reduced penetrance. Several known hereditary syndromes or genes are associated with an increased risk of developing PC and account for approximately 2% of PCs. These syndromes include the hereditary breast-ovarian cancer syndrome, Peutz-Jeghers syndrome, familial atypical multiple mole melanoma, Lynch syndrome, familial polyposis, ataxia-telangiectasia, and hereditary pancreatitis. Appropriate screening using methods such as biomarkers or imaging, with endoscopic ultrasound and magnetic resonance imaging, may assist in the early detection of neoplastic lesions in the high-risk population. If these lesions are detected and treated before the development of invasive carcinoma, PC disease morbidity and mortality may be improved. This review will focus on familial PC and other hereditary syndromes implicated in the increased risk of PC; it will also highlight current screening methods and the future of new screening modalities.
Collapse
Affiliation(s)
- Sara Welinsky
- Samuel F. Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aimee L Lucas
- Samuel F. Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Lipner MB, Yeh JJ. Sequencing Pancreatic Juice: Squeezing the Most Out of Surveillance. Clin Cancer Res 2018; 24:2713-2715. [PMID: 29500277 DOI: 10.1158/1078-0432.ccr-18-0215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/19/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022]
Abstract
Next-generation sequencing of pancreatic juice can detect and quantify tumor-promoting mutations, supporting imaging and cytology findings to predict the degree of dysplasia in patients at high risk for pancreatic cancer. Future studies are needed to optimize this approach and determine how it best fits into clinical practice. Clin Cancer Res; 24(12); 2713-5. ©2018 AACRSee related article by Suenaga et al., p. 2963.
Collapse
Affiliation(s)
- Matthew B Lipner
- Department of Pharmacology, School of Medicine, The University of North Carolina, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, North Carolina
| | - Jen Jen Yeh
- Department of Pharmacology, School of Medicine, The University of North Carolina, Chapel Hill, North Carolina. .,Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, North Carolina.,Department of Surgery, Division of Surgical Oncology and Endocrinology, The University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
31
|
Felsenstein M, Hruban RH, Wood LD. New Developments in the Molecular Mechanisms of Pancreatic Tumorigenesis. Adv Anat Pathol 2018; 25:131-142. [PMID: 28914620 DOI: 10.1097/pap.0000000000000172] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Pancreatic cancer is an aggressive disease with a dismal prognosis in dire need of novel diagnostic and therapeutic approaches. The past decade has witnessed an explosion of data on the genetic alterations that occur in pancreatic cancer, as comprehensive next-generation sequencing analyses have been performed on samples from large cohorts of patients. These studies have defined the genomic landscape of this disease and identified novel candidates whose mutations contribute to pancreatic tumorigenesis. They have also clarified the genetic alterations that underlie multistep tumorigenesis in precursor lesions and provided insights into clonal evolution in pancreatic neoplasia. In addition to these important insights into pancreatic cancer biology, these large scale genomic studies have also provided a foundation for the development of novel early detection strategies and targeted therapies. In this review, we discuss the results of these comprehensive sequencing studies of pancreatic neoplasms, with a particular focus on how their results will impact the clinical care of patients with pancreatic cancer.
Collapse
|
32
|
Validation of Biomarkers for Early Detection of Pancreatic Cancer: Summary of The Alliance of Pancreatic Cancer Consortia for Biomarkers for Early Detection Workshop. Pancreas 2018; 47:135-141. [PMID: 29346214 PMCID: PMC5777224 DOI: 10.1097/mpa.0000000000000973] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is the third leading cause of cancer death in the United States, and the 5-year relative survival for patients diagnosed with pancreatic cancer is less than 10%. Early intervention is the key to a better survival outcome. Currently, there are no biomarkers that can reliably detect pancreatic cancer at an early stage or identify precursors that are destined to progress to malignancy. The National Cancer Institute in partnership with the Kenner Family Research Fund and the Pancreatic Cancer Action Network convened a Data Jamboree on Biomarkers workshop on December 5, 2016, to discuss and evaluate existing or newly developed biomarkers and imaging methods for early detection of pancreatic cancer. The primary goal of this workshop was to determine if there are any promising biomarkers for early detection of pancreatic cancer that are ready for clinical validation. The Alliance of Pancreatic Cancer Consortia for Biomarkers for Early Detection, formed under the auspices of this workshop, will provide the common platform and the resources necessary for validation. Although none of the biomarkers evaluated seemed ready for a large-scale biomarker validation trial, a number of them had sufficiently high sensitivity and specificity to warrant additional research, especially if combined with other biomarkers to form a panel.
Collapse
|
33
|
Suenaga M, Yu J, Shindo K, Tamura K, Almario JA, Zaykoski C, Witmer PD, Fesharakizadeh S, Borges M, Lennon AM, Shin EJ, Canto MI, Goggins M. Pancreatic Juice Mutation Concentrations Can Help Predict the Grade of Dysplasia in Patients Undergoing Pancreatic Surveillance. Clin Cancer Res 2018; 24:2963-2974. [PMID: 29301828 DOI: 10.1158/1078-0432.ccr-17-2463] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/15/2017] [Accepted: 12/29/2017] [Indexed: 12/21/2022]
Abstract
Purpose: The measurement of mutations in pancreatic juice samples collected from the duodenum during endoscopic ultrasound (EUS) may improve the diagnostic evaluation of patients undergoing pancreatic surveillance. Our aim was to evaluate the accuracy of using pancreatic juice mutation concentrations to predict the presence and histologic grade of neoplasia in the pancreas.Experimental Design: Digital next-generation sequencing (NGS) of pancreatic juice DNA using a targeted 12-gene panel was performed on 67 patients undergoing pancreatic evaluation during EUS, including patients with pancreatic ductal adenocarcinoma, patients who subsequently underwent pancreatic resection for precursor lesions, patients undergoing surveillance for their familial/inherited susceptibility to pancreatic cancer, and normal pancreas disease controls.Results: Patients with pancreatic cancer or high-grade dysplasia as their highest grade lesion had significantly higher pancreatic juice mutation concentrations than all other subjects (mean/SD digital NGS score; 46.6 ± 69.7 vs. 6.2 ± 11.6, P = 0.02). Pancreatic juice mutation concentrations distinguished patients with pancreatic cancer or high-grade dysplasia in their resection specimen from all other subjects with 72.2% sensitivity and 89.4% specificity [area under the curve (AUC) = 0.872]. Mutant TP53/SMAD4 concentrations could distinguish patients with pancreatic cancer or high-grade dysplasia in their resection specimen from all other subjects with 61.1% sensitivity and 95.7% specificity (AUC = 0.819). Among 31 high-risk individuals under surveillance, 2 of the 3 individuals with most abnormal pancreatic juice mutation profiles also had the most abnormalities on pancreatic imaging.Conclusions: Pancreatic juice mutation analysis using digital NGS has potential diagnostic utility in the evaluation of patients undergoing pancreatic surveillance. Clin Cancer Res; 24(12); 2963-74. ©2018 AACRSee related commentary by Lipner and Yeh, p. 2713.
Collapse
Affiliation(s)
- Masaya Suenaga
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jun Yu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Koji Shindo
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Koji Tamura
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Jose Alejandro Almario
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Christopher Zaykoski
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - P Dane Witmer
- Center for Inherited Disease Research (CIDR), Baltimore, Maryland
| | - Shahriar Fesharakizadeh
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Anne-Marie Lennon
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland.,Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Eun-Ji Shin
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Marcia Irene Canto
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland. .,Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
34
|
Kong B, Bruns P, Behler NA, Chang L, Schlitter AM, Cao J, Gewies A, Ruland J, Fritzsche S, Valkovskaya N, Jian Z, Regel I, Raulefs S, Irmler M, Beckers J, Friess H, Erkan M, Mueller NS, Roth S, Hackert T, Esposito I, Theis FJ, Kleeff J, Michalski CW. Dynamic landscape of pancreatic carcinogenesis reveals early molecular networks of malignancy. Gut 2018; 67:146-156. [PMID: 27646934 DOI: 10.1136/gutjnl-2015-310913] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 08/15/2016] [Accepted: 08/23/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The initial steps of pancreatic regeneration versus carcinogenesis are insufficiently understood. Although a combination of oncogenic Kras and inflammation has been shown to induce malignancy, molecular networks of early carcinogenesis remain poorly defined. DESIGN We compared early events during inflammation, regeneration and carcinogenesis on histological and transcriptional levels with a high temporal resolution using a well-established mouse model of pancreatitis and of inflammation-accelerated KrasG12D-driven pancreatic ductal adenocarcinoma. Quantitative expression data were analysed and extensively modelled in silico. RESULTS We defined three distinctive phases-termed inflammation, regeneration and refinement-following induction of moderate acute pancreatitis in wild-type mice. These corresponded to different waves of proliferation of mesenchymal, progenitor-like and acinar cells. Pancreas regeneration required a coordinated transition of proliferation between progenitor-like and acinar cells. In mice harbouring an oncogenic Kras mutation and challenged with pancreatitis, there was an extended inflammatory phase and a parallel, continuous proliferation of mesenchymal, progenitor-like and acinar cells. Analysis of high-resolution transcriptional data from wild-type animals revealed that organ regeneration relied on a complex interaction of a gene network that normally governs acinar cell homeostasis, exocrine specification and intercellular signalling. In mice with oncogenic Kras, a specific carcinogenic signature was found, which was preserved in full-blown mouse pancreas cancer. CONCLUSIONS These data define a transcriptional signature of early pancreatic carcinogenesis and a molecular network driving formation of preneoplastic lesions, which allows for more targeted biomarker development in order to detect cancer earlier in patients with pancreatitis.
Collapse
Affiliation(s)
- Bo Kong
- Department of Surgery, Technische Universität München (TUM), Munich, Germany.,Department of Gastroenterology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, China
| | - Philipp Bruns
- Department of Surgery, Technische Universität München (TUM), Munich, Germany.,Institute of Computational Biology, Helmholtz-Zentrum München GmbH, Neuherberg, Germany
| | - Nora A Behler
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | - Ligong Chang
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | | | - Jing Cao
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | - Andreas Gewies
- Institute für Klinische Chemie und Pathobiochemie, TUM, Munich, Germany.,Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Neuherberg, Germany.,German Cancer Consortium (DKTK) at the partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Ruland
- Institute für Klinische Chemie und Pathobiochemie, TUM, Munich, Germany.,German Cancer Consortium (DKTK) at the partner site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Sina Fritzsche
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | | | - Ziying Jian
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | - Ivonne Regel
- Institute of Pathology, Heinrich-Heine University, Duesseldorf, Germany
| | - Susanne Raulefs
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, Neuherberg, Germany.,Chair of Experimental Genetics, Technische Universität München, Freising, Germany.,Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Helmut Friess
- Department of Surgery, Technische Universität München (TUM), Munich, Germany
| | - Mert Erkan
- Department of Surgery, Koc University, Istanbul, Turkey
| | - Nikola S Mueller
- Institute of Computational Biology, Helmholtz-Zentrum München GmbH, Neuherberg, Germany
| | - Susanne Roth
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - Irene Esposito
- Institute of Pathology, Heinrich-Heine University, Duesseldorf, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz-Zentrum München GmbH, Neuherberg, Germany.,Department of Mathematics, TUM, Munich, Germany
| | - Jörg Kleeff
- Department of Surgery, Technische Universität München (TUM), Munich, Germany.,NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
35
|
Abstract
OBJECTIVE Secretin-stimulated pancreatic juice is collected from the duodenum and analyzed to identify biomarkers of pancreatic neoplasia, but the optimal duration of pancreatic juice collection is not known. METHODS We compared the yield of KRAS mutations detected in pancreatic juice samples aspirated from near the duodenal papilla at 1 to 5, 6 to 10, and 11 to 15 minutes after secretin infusion, and from the third part of the duodenum (at 15 minutes) from 45 patients undergoing endoscopic ultrasound pancreatic surveillance. KRAS mutation concentrations were measured by using droplet digital polymerase chain reaction. RESULTS Forty of 45 patients had KRAS mutations detected in their pancreatic juice, and most patients' juice samples had more than 1 KRAS mutation. Of 106 KRAS mutations detected in 171 pancreatic juice samples, 58 were detected in the 5-minute samples, 70 mutations were detected in the 10-minute samples, and 65 were detected in the 15-minute samples. Nine patients who did not have KRAS mutations detected in their 5-minute sample had mutations detected in samples collected at later time points. Ninety-percent of all pancreatic juice mutations detected in any sample were detected in the 5- or 10-minute samples. CONCLUSIONS Collecting pancreatic juice for 10 minutes after secretin infusion increases the likelihood of detecting pancreatic juice mutations over shorter collections.
Collapse
|
36
|
Farrell JJ. Intraductal papillary mucinous neoplasm to pancreas ductal adenocarcinoma sequence and pancreas cancer screening. Endosc Ultrasound 2018; 7:314-318. [PMID: 30323160 PMCID: PMC6199903 DOI: 10.4103/eus.eus_49_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- James J Farrell
- Department of Digestive Diseases, Yale Center for Pancreatic Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
37
|
Pittman ME, Rao R, Hruban RH. Classification, Morphology, Molecular Pathogenesis, and Outcome of Premalignant Lesions of the Pancreas. Arch Pathol Lab Med 2017; 141:1606-1614. [PMID: 29189063 DOI: 10.5858/arpa.2016-0426-ra] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT - Invasive pancreatic ductal adenocarcinoma has a greater than 90% mortality rate at 5 years. Understanding noninvasive, curable precursor lesions gives us the best hope for reducing mortality from pancreatic ductal adenocarcinoma. The 3 pancreatic precursor lesions that have been well studied include intraductal papillary mucinous neoplasm, mucinous cystic neoplasm, and pancreatic intraepithelial neoplasia. OBJECTIVE - To give an update on the latest clinical, molecular, and pathologic advances in intraductal papillary mucinous neoplasm, mucinous cystic neoplasm, and pancreatic intraepithelial neoplasia for the general surgical pathologist. DATA SOURCES - The current literature was analyzed and the authors' experiences with institutional and consult material were incorporated. CONCLUSIONS - Our understanding of the molecular alterations that lead from pancreatic precursor lesion to invasive carcinoma continues to evolve. These advances aid clinicians in their treatment decisions and researchers in their search for actionable, druggable targets.
Collapse
|
38
|
Can we better predict the biologic behavior of incidental IPMN? A comprehensive analysis of molecular diagnostics and biomarkers in intraductal papillary mucinous neoplasms of the pancreas. Langenbecks Arch Surg 2017; 403:151-194. [DOI: 10.1007/s00423-017-1644-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
|
39
|
How to suction pancreatic juice from the duodenum: Endoscope, catheter, or cap-assisted? Gastrointest Endosc 2017; 86:1157-1159. [PMID: 29146085 DOI: 10.1016/j.gie.2017.03.1516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/18/2017] [Indexed: 12/11/2022]
|
40
|
Suenaga M, Sadakari Y, Almario JA, Borges M, Lennon AM, Shin EJ, Canto MI, Goggins M. Using an endoscopic distal cap to collect pancreatic fluid from the ampulla (with video). Gastrointest Endosc 2017; 86:1152-1156.e2. [PMID: 28259593 PMCID: PMC5581309 DOI: 10.1016/j.gie.2017.02.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Duodenal collections of pancreatic fluid can be used as a source of mutations and other markers of pancreatic ductal neoplasia, but admixing pancreatic juice with duodenal contents lowers the concentrations of mutations. Collecting pancreatic fluid directly from the ampulla could yield a purer sample of pancreatic fluid. METHODS We used an endoscopic distal cap attachment to "cap" the ampulla and collect secretin-stimulated pancreatic fluid samples for 5 minutes from 81 patients undergoing pancreatic evaluation as part of the Cancer of the Pancreas Screening studies. We compared mutation concentrations (K-ras and GNAS) measured by droplet-digital PCR (ddPCR) in "cap-collected juice" samples to those found in juice samples obtained from 77 patients collected by aspiration from the duodenal lumen without capping the ampulla. RESULTS Among all subjects, mutation concentrations were higher in pancreatic juice samples collected using the endoscopic cap method (median, .028%; IQR, 0-.077) compared with the noncap-collected (median, .019%; IQR, 0-.044; P = .055). Among pancreatic juice samples with detectable mutations, mutation concentrations were higher in the cap-collected juice samples than in those collected without the cap (.055%; IQR, .026-.092 vs .032%; IQR, .020-.066; P = .031). CONCLUSIONS Collecting pancreatic juice directly from the ampulla using an endoscopic distal cap yields higher concentrations of pancreatic fluid mutations.
Collapse
Affiliation(s)
- Masaya Suenaga
- Department of Pathology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Yoshihiko Sadakari
- Department of Pathology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Jose Alejandro Almario
- Department of Pathology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Anne-Marie Lennon
- Department of Medicine, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
- Department of Surgery, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Eun-Ji Shin
- Department of Medicine, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Marcia Irene Canto
- Department of Medicine, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
- Department of Oncology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
- Department of Medicine, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
- Department of Oncology, The Sol Goldman Pancreatic, Cancer Research Center, Johns Hopkins Medical Institutions
| |
Collapse
|
41
|
Basar O, Brugge WR. My Treatment Approach: Pancreatic Cysts. Mayo Clin Proc 2017; 92:1519-1531. [PMID: 28890216 DOI: 10.1016/j.mayocp.2017.06.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/21/2017] [Accepted: 06/06/2017] [Indexed: 12/12/2022]
Abstract
Our treatment approach for either symptomatic or incidentally found pancreatic cysts continues to improve. The true incidence of pancreatic cysts is not known, and pancreatic cystic neoplasms, especially intraductal papillary mucinous neoplasms, are currently most commonly diagnosed and resected. This is a result of increasing awareness, widespread availability of imaging, and better understanding of the nature of pancreatic cysts as well. Recent studies on molecular analysis and devices such as microbiopsy forceps help us better define and select the treatment approach to alleviate symptoms and to prevent malignant tumors while avoiding unnecessary surgery.
Collapse
Affiliation(s)
- Omer Basar
- Pancreas Biliary Center, Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA.
| | - William R Brugge
- Pancreas Biliary Center, Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| |
Collapse
|
42
|
Yu J, Sadakari Y, Shindo K, Suenaga M, Brant A, Almario JAN, Borges M, Barkley T, Fesharakizadeh S, Ford M, Hruban RH, Shin EJ, Lennon AM, Canto MI, Goggins M. Digital next-generation sequencing identifies low-abundance mutations in pancreatic juice samples collected from the duodenum of patients with pancreatic cancer and intraductal papillary mucinous neoplasms. Gut 2017; 66:1677-1687. [PMID: 27432539 PMCID: PMC5243915 DOI: 10.1136/gutjnl-2015-311166] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 04/27/2016] [Accepted: 05/19/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Secretin-stimulated pancreatic juice contains DNA shed from cells lining the pancreatic ducts. Genetic analysis of this fluid may form a test to detect pancreatic ductal neoplasia. DESIGN We employed digital next-generation sequencing ('digital NGS') to detect low-abundance mutations in secretin-stimulated juice samples collected from the duodenum of subjects enrolled in Cancer of the Pancreas Screening studies at Johns Hopkins Hospital. For each juice sample, digital NGS necessitated 96 NGS reactions sequencing nine genes. The study population included 115 subjects (53 discovery, 62 validation) (1) with pancreatic ductal adenocarcinoma (PDAC), (2) intraductal papillary mucinous neoplasm (IPMN), (3) controls with non-suspicious pancreata. RESULTS Cases with PDAC and IPMN were more likely to have mutant DNA detected in pancreatic juice than controls (both p<0.0001); mutant DNA concentrations were higher in patients with PDAC than IPMN (p=0.003) or controls (p<0.001). TP53 and/or SMAD4 mutations were commonly detected in juice samples from patients with PDAC and were not detected in controls (p<0.0001); mutant TP53/SMAD4 concentrations could distinguish PDAC from IPMN cases with 32.4% sensitivity, 100% specificity (area under the curve, AUC 0.73, p=0.0002) and controls (AUC 0.82, p<0.0001). Two of four patients who developed pancreatic cancer despite close surveillance had SMAD4/TP53 mutations from their cancer detected in juice samples collected over 1 year prior to their pancreatic cancer diagnosis when no suspicious pancreatic lesions were detected by imaging. CONCLUSIONS The detection in pancreatic juice of mutations important for the progression of low-grade dysplasia to high-grade dysplasia and invasive pancreatic cancer may improve the management of patients undergoing pancreatic screening and surveillance.
Collapse
Affiliation(s)
- Jun Yu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yoshihiko Sadakari
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Koji Shindo
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Masaya Suenaga
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aaron Brant
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jose Alejandro Navarro Almario
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Borges
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas Barkley
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shahriar Fesharakizadeh
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Madeline Ford
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eun Ji Shin
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anne Marie Lennon
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcia Irene Canto
- Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
43
|
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
|
44
|
Sheth SG, Conwell DL, Whitcomb DC, Alsante M, Anderson MA, Barkin J, Brand R, Cote GA, Freedman SD, Gelrud A, Gorelick F, Lee LS, Morgan K, Pandol S, Singh VK, Yadav D, Wilcox CM, Hart PA. Academic Pancreas Centers of Excellence: Guidance from a multidisciplinary chronic pancreatitis working group at PancreasFest. Pancreatology 2017; 17:419-430. [PMID: 28268158 PMCID: PMC5525332 DOI: 10.1016/j.pan.2017.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/22/2017] [Accepted: 02/26/2017] [Indexed: 12/11/2022]
Abstract
Chronic pancreatitis (CP) is a progressive inflammatory disease, which leads to loss of pancreatic function and other disease-related morbidities. A group of academic physicians and scientists developed comprehensive guidance statements regarding the management of CP that include its epidemiology, diagnosis, medical treatment, surgical treatment, and screening. The statements were developed through literature review, deliberation, and consensus opinion. These statements were ultimately used to develop a conceptual framework for the multidisciplinary management of chronic pancreatitis referred to as an academic pancreas center of excellence (APCOE).
Collapse
Affiliation(s)
- Sunil G Sheth
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Darwin L Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - David C Whitcomb
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh and UPMC, Pittsburgh, PA, United States
| | | | - Michelle A Anderson
- Division of Gastroenterology, University of Michigan Hospital and Health Systems, Ann Arbor, MI, United States
| | - Jamie Barkin
- University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Randall Brand
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh and UPMC, Pittsburgh, PA, United States
| | - Gregory A Cote
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC, United States
| | - Steven D Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Andres Gelrud
- Center for Endoscopic Research and Therapeutics, University of Chicago, Chicago, IL, United States
| | - Fred Gorelick
- Section of Digestive Diseases, Yale University and VA Healthcare, West Haven, CT, United States
| | - Linda S Lee
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Boston, MA, United States
| | - Katherine Morgan
- Division of Gastrointestinal and Laparoscopic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Pandol
- Division of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Vikesh K Singh
- Division of Gastroenterology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh and UPMC, Pittsburgh, PA, United States
| | - C Mel Wilcox
- Division of Gastroenterology, University of Alabama, Birmingham, AL, United States
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, OH, United States.
| |
Collapse
|
45
|
Hosoda W, Chianchiano P, Griffin JF, Pittman ME, Brosens LA, Noë M, Yu J, Shindo K, Suenaga M, Rezaee N, Yonescu R, Ning Y, Albores-Saavedra J, Yoshizawa N, Harada K, Yoshizawa A, Hanada K, Yonehara S, Shimizu M, Uehara T, Samra JS, Gill AJ, Wolfgang CL, Goggins MG, Hruban RH, Wood LD. Genetic analyses of isolated high-grade pancreatic intraepithelial neoplasia (HG-PanIN) reveal paucity of alterations in TP53 and SMAD4. J Pathol 2017; 242:16-23. [PMID: 28188630 DOI: 10.1002/path.4884] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/13/2017] [Accepted: 01/30/2017] [Indexed: 12/28/2022]
Abstract
High-grade pancreatic intraepithelial neoplasia (HG-PanIN) is the major precursor of pancreatic ductal adenocarcinoma (PDAC) and is an ideal target for early detection. To characterize pure HG-PanIN, we analysed 23 isolated HG-PanIN lesions occurring in the absence of PDAC. Whole-exome sequencing of five of these HG-PanIN lesions revealed a median of 33 somatic mutations per lesion, with a total of 318 mutated genes. Targeted next-generation sequencing of 17 HG-PanIN lesions identified KRAS mutations in 94% of the lesions. CDKN2A alterations occurred in six HG-PanIN lesions, and RNF43 alterations in five. Mutations in TP53, GNAS, ARID1A, PIK3CA, and TGFBR2 were limited to one or two HG-PanINs. No non-synonymous mutations in SMAD4 were detected. Immunohistochemistry for p53 and SMAD4 proteins in 18 HG-PanINs confirmed the paucity of alterations in these genes, with aberrant p53 labelling noted only in three lesions, two of which were found to be wild type in sequencing analyses. Sixteen adjacent LG-PanIN lesions from ten patients were also sequenced using targeted sequencing. LG-PanIN harboured KRAS mutations in 94% of the lesions; mutations in CDKN2A, TP53, and SMAD4 were not identified. These results suggest that inactivation of TP53 and SMAD4 are late genetic alterations, predominantly occurring in invasive PDAC. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Waki Hosoda
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter Chianchiano
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James F Griffin
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Meredith E Pittman
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Lodewijk Aa Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michaël Noë
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jun Yu
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Koji Shindo
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Masaya Suenaga
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Neda Rezaee
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Raluca Yonescu
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yi Ning
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Naohiko Yoshizawa
- The First Department of Internal Medicine, Mie University School of Medicine, Tsu, Japan
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Keiji Hanada
- Center for Gastroendoscopy, Onomichi General Hospital, Onomichi, Japan
| | - Shuji Yonehara
- Department of Pathology, Onomichi General Hospital, Onomich, Japan
| | - Michio Shimizu
- Diagnostic Pathology Center, Hakujikai Memorial Hospital, Tokyo, Japan
| | - Takeshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Jaswinder S Samra
- Department of Gastrointestinal Surgery, Royal North Shore Hospital and Discipline of Surgery, University of Sydney, Sydney, Australia
| | - Anthony J Gill
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research Royal North Shore Hospital and University of Sydney, Sydney, Australia
| | - Christopher L Wolfgang
- Department of Surgery, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael G Goggins
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Medicine, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laura D Wood
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Portal Venous Blood Circulation Supports Immunosuppressive Environment and Pancreatic Cancer Circulating Tumor Cell Activation. Pancreas 2017; 46:116-123. [PMID: 27400259 DOI: 10.1097/mpa.0000000000000667] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Aggressive spread and liver metastases are predominant features of pancreatic ductal adenocarcinoma (PDAC). This study investigates activation of PDAC circulating tumor cells (CTC) and immunosuppression in the portal venous system. METHODS Portal venous and peripheral blood were collected during pancreaticoduodenectomy from patients with PDAC (n = 21) or other non-PDAC pancreatic conditions (n = 20). Circulating tumor cells were isolated by fluorescence-activated cell sorting and characterized for messenger RNA (mRNA) expression and acetylated chromatin encoding K-RAS exon 12 mutation (K-RASmut). Myeloid-derived suppressor cells (MDSC) were identified using flow cytometry. RESULTS Pancreatic ductal adenocarcinoma K-RASmut mRNA expression in portal venous blood CTC was significantly elevated compared with preoperative and postoperative peripheral blood (P = 0.0123 and P = 0.0246, respectively). There was no significant variation in total CTC numbers between portal and peripheral blood.Portal venous M-MDSC were elevated compared with peripheral blood in PDAC patients (P = 0.0065). M-MDSC increases correlated with K-RASmut mRNA-expressing CTC present in PDAC portal blood (P < 0.0001). CONCLUSIONS Association of MDSC with active CTC in portal venous blood may support immunosuppression within the portal venous circulation to promote PDAC CTC survival.
Collapse
|
47
|
Overbeek KA, Cahen DL, Canto MI, Bruno MJ. Surveillance for neoplasia in the pancreas. Best Pract Res Clin Gastroenterol 2016; 30:971-986. [PMID: 27938791 PMCID: PMC5552042 DOI: 10.1016/j.bpg.2016.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 10/25/2016] [Accepted: 10/31/2016] [Indexed: 01/31/2023]
Abstract
Despite its low incidence in the general population, pancreatic cancer is one of the leading causes of cancer-related mortality. Survival greatly depends on operability, but most patients present with unresectable disease. Therefore, there is great interest in the early detection of pancreatic cancer and its precursor lesions by surveillance. Worldwide, several programs have been initiated for individuals at high risk for pancreatic cancer. Their first results suggest that surveillance in high-risk individuals is feasible, but their effectiveness in decreasing mortality remains to be proven. This review will discuss which individuals are eligible for surveillance, which lesions are aimed to be detected, and which surveillance modalities are being used in current clinical practice. Furthermore, it addresses the management of abnormalities found during surveillance and topics for future research.
Collapse
Affiliation(s)
- Kasper A. Overbeek
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, ‘s Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands,Corresponding author. Fax: +31 10 703 03 31
| | - Djuna L. Cahen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, ‘s Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Marcia Irene Canto
- Division of Gastroenterology and Hepatology, The Johns Hopkins Medical Institutions, 1800 Orleans St., Blalock 407, Baltimore, MD, 21287, USA
| | - Marco J. Bruno
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, ‘s Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| |
Collapse
|
48
|
Takai E, Yachida S. Circulating tumor DNA as a liquid biopsy target for detection of pancreatic cancer. World J Gastroenterol 2016; 22:8480-8488. [PMID: 27784960 PMCID: PMC5064029 DOI: 10.3748/wjg.v22.i38.8480] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/30/2016] [Accepted: 08/05/2016] [Indexed: 02/06/2023] Open
Abstract
Most pancreatic cancer patients present with advanced metastatic disease, resulting in extremely poor 5-year survival, mainly because of the lack of a reliable modality for early detection and limited therapeutic options for advanced disease. Therefore, there is a need for minimally-invasive diagnostic tools for detecting pancreatic cancer at an early stage, when curative surgery and also novel therapeutic approaches including precision medicine may be feasible. The “liquid biopsy” addresses these unmet clinical needs based on the concept that simple peripheral blood sampling and detection of circulating tumor DNA (ctDNA) could provide diagnostic information. In this review, we provide an overview of the current status of blood-based tests for diagnosis of pancreatic cancer and the potential utility of ctDNA for precision medicine. We also discuss challenges that remain to be addressed in developing practical ctDNA-based liquid biopsy approaches for early diagnosis of pancreatic cancer.
Collapse
|
49
|
Hart PA, Topazian M, Raimondo M, Cruz-Monserrate Z, Fisher WE, Lesinski GB, Steen H, Conwell DL. Endoscopic Pancreas Fluid Collection: Methods and Relevance for Clinical Care and Translational Science. Am J Gastroenterol 2016; 111:1258-66. [PMID: 27481304 PMCID: PMC5568003 DOI: 10.1038/ajg.2016.297] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/01/2016] [Indexed: 02/08/2023]
Abstract
Pancreatic secretions have an important role in the regulation of a normal nutritional state but can be altered owing to a variety of pathophysiological mechanisms in the context of exocrine pancreatic disease. The development of an endoscopic technique for collection of pancreatic fluid, termed endoscopic pancreatic function testing, has led to improved understanding of these alterations and is particularly helpful to characterize chronic pancreatitis. In addition, investigators have found endoscopically collected pancreatic fluid to be a valuable biofluid for the purposes of translational science. Techniques such as proteomic, cytokine, genetic mutation, DNA methylation, and microRNA analyses, among others, can be utilized to gain a better understanding of the molecular characteristics of chronic pancreatitis and other pancreatic diseases. Endoscopic collection of pancreatic fluid is safe and relatively straightforward, permitting opportunities for longitudinal analysis of these translational markers throughout the course of disease. This manuscript summarizes our current knowledge of pancreatic fluid, with an emphasis on proper techniques for sample collection and handling, its clinical utility, and preliminary observations in translational science.
Collapse
Affiliation(s)
- Phil A. Hart
- Section of Pancreatic Disorders, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Mark Topazian
- Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Massimo Raimondo
- Division of Gastroenterology and Hepatology, Mayo Clinic Jacksonville, Jacksonville, Florida, USA
| | - Zobeida Cruz-Monserrate
- Section of Pancreatic Disorders, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - William E. Fisher
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Gregory B. Lesinski
- Section of Pancreatic Disorders, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Hanno Steen
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Msaachusetts, USA
| | - Darwin L. Conwell
- Section of Pancreatic Disorders, Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
50
|
Abstract
Pancreatic cancer is the fourth leading cause of cancer related deaths in the United States with a 5-year survival rate of less than 10%. The Division of Cancer Prevention of the National Cancer Institute sponsored the Pancreatic Cancer Chemoprevention Translational Workshop on September 10 to 11, 2015. The goal of the workshop was to obtain information regarding the current state of the science and future scientific areas that should be prioritized for pancreatic cancer prevention research, including early detection and intervention for high-risk precancerous lesions. The workshop addressed the molecular/genetic landscape of pancreatic cancer and precursor lesions, high-risk populations and criteria to identify a high-risk population for potential chemoprevention trials, identification of chemopreventative/immunopreventative agents, and use of potential biomarkers and imaging for assessing short-term efficacy of a preventative agent. The field of chemoprevention for pancreatic cancer is emerging, and this workshop was organized to begin to address these important issues and promote multi-institutional efforts in this area. The meeting participants recommended the development of an National Cancer Institute working group to coordinate efforts, provide a framework, and identify opportunities for chemoprevention of pancreatic cancer.
Collapse
|