Skip to main content
Top
Published in: BMC Cancer 1/2019

Open Access 01-12-2019 | Pancreatic Cancer | Research article

Mutation analysis by deep sequencing of pancreatic juice from patients with pancreatic ductal adenocarcinoma

Authors: Man Hung Choi, Eline Mejlænder-Andersen, Sophia Manueldas, Khadija El Jellas, Solrun J. Steine, Kjersti Tjensvoll, Hege Aase Sætran, Stian Knappskog, Dag Hoem, Oddmund Nordgård, Randi Hovland, Anders Molven

Published in: BMC Cancer | Issue 1/2019

Login to get access

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.
Appendix
Available only for authorised users
Literature
2.
go back to reference Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. 2014;371:1039–49.CrossRef Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med. 2014;371:1039–49.CrossRef
3.
go back to reference Chari ST, Kelly K, Hollingsworth MA, Thayer SP, Ahlquist DA, Andersen DK, et al. Early detection of sporadic pancreatic cancer: summative review. Pancreas. 2015;44:693–712.CrossRef Chari ST, Kelly K, Hollingsworth MA, Thayer SP, Ahlquist DA, Andersen DK, et al. Early detection of sporadic pancreatic cancer: summative review. Pancreas. 2015;44:693–712.CrossRef
4.
go back to reference Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–38.CrossRef Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–38.CrossRef
5.
go back to reference Lipner MB, Yeh JJ. Sequencing pancreatic juice: squeezing the most out of surveillance. Clin Cancer Res. 2018. Lipner MB, Yeh JJ. Sequencing pancreatic juice: squeezing the most out of surveillance. Clin Cancer Res. 2018.
6.
go back to reference Oldfield LE, Connor AA, Gallinger S. Molecular events in the natural history of pancreatic cancer. Trends Cancer. 2017;3:336–46.CrossRef Oldfield LE, Connor AA, Gallinger S. Molecular events in the natural history of pancreatic cancer. Trends Cancer. 2017;3:336–46.CrossRef
7.
go back to reference Murphy SJ, Hart SN, Lima JF, Kipp BR, Klebig M, Winters JL, et al. Genetic alterations associated with progression from pancreatic intraepithelial neoplasia to invasive pancreatic tumor. Gastroenterology. 2013;145:1098–109 e1.CrossRef Murphy SJ, Hart SN, Lima JF, Kipp BR, Klebig M, Winters JL, et al. Genetic alterations associated with progression from pancreatic intraepithelial neoplasia to invasive pancreatic tumor. Gastroenterology. 2013;145:1098–109 e1.CrossRef
8.
go back to reference The Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32:185–203.e13. The Cancer Genome Atlas Research Network. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell. 2017;32:185–203.e13.
9.
go back to reference Shi C, Fukushima N, Abe T, Bian Y, Hua L, Wendelburg BJ, et al. Sensitive and quantitative detection of KRAS2 gene mutations in pancreatic duct juice differentiates patients with pancreatic cancer from chronic pancreatitis, potential for early detection. Cancer Biology & Therapy. 2008;7:353–60.CrossRef Shi C, Fukushima N, Abe T, Bian Y, Hua L, Wendelburg BJ, et al. Sensitive and quantitative detection of KRAS2 gene mutations in pancreatic duct juice differentiates patients with pancreatic cancer from chronic pancreatitis, potential for early detection. Cancer Biology & Therapy. 2008;7:353–60.CrossRef
10.
go back to reference Kanda M, Knight S, Topazian M, Syngal S, Farrell J, Lee J, et al. Mutant GNAS detected in duodenal collections of secretin-stimulated pancreatic juice indicates the presence or emergence of pancreatic cysts. Gut. 2013;62:1024–33.CrossRef Kanda M, Knight S, Topazian M, Syngal S, Farrell J, Lee J, et al. Mutant GNAS detected in duodenal collections of secretin-stimulated pancreatic juice indicates the presence or emergence of pancreatic cysts. Gut. 2013;62:1024–33.CrossRef
11.
go back to reference Kanda M, Sadakari Y, Borges M, Topazian M, Farrell J, Syngal S, et al. Mutant TP53 in duodenal samples of pancreatic juice from patients with pancreatic cancer or high-grade dysplasia. Clinical Gastroenterology and Hepatology . 2013;11:719–730 e5. Kanda M, Sadakari Y, Borges M, Topazian M, Farrell J, Syngal S, et al. Mutant TP53 in duodenal samples of pancreatic juice from patients with pancreatic cancer or high-grade dysplasia. Clinical Gastroenterology and Hepatology . 2013;11:719–730 e5.
12.
go back to reference Eshleman JR, Norris AL, Sadakari Y, Debeljak M, Borges M, Harrington C, et al. KRAS and guanine nucleotide-binding protein mutations in pancreatic juice collected from the duodenum of patients at high risk for neoplasia undergoing endoscopic ultrasound. Clin Gastroenterol Hepatol. 2015;13:963–9 e4.CrossRef Eshleman JR, Norris AL, Sadakari Y, Debeljak M, Borges M, Harrington C, et al. KRAS and guanine nucleotide-binding protein mutations in pancreatic juice collected from the duodenum of patients at high risk for neoplasia undergoing endoscopic ultrasound. Clin Gastroenterol Hepatol. 2015;13:963–9 e4.CrossRef
13.
go back to reference Yu J, Sadakari Y, Shindo K, Suenaga M, Brant A, Almario JAN, et al. 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;66:1677–87.CrossRef Yu J, Sadakari Y, Shindo K, Suenaga M, Brant A, Almario JAN, et al. 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;66:1677–87.CrossRef
14.
go back to reference Suenaga M, Yu J, Shindo K, Tamura K, Almario JAN, Zaykoski CM, et al. Pancreatic juice mutation concentrations can help predict the grade of dysplasia in patients undergoing pancreatic surveillance. Clin Cancer Res. 2018;24:2963–74.CrossRef Suenaga M, Yu J, Shindo K, Tamura K, Almario JAN, Zaykoski CM, et al. Pancreatic juice mutation concentrations can help predict the grade of dysplasia in patients undergoing pancreatic surveillance. Clin Cancer Res. 2018;24:2963–74.CrossRef
16.
go back to reference Li Q, Wang K. InterVar: clinical interpretation of genetic variants by the 2015 ACMG-AMP guidelines. Am J Hum Genet. 2017;100:267–80.CrossRef Li Q, Wang K. InterVar: clinical interpretation of genetic variants by the 2015 ACMG-AMP guidelines. Am J Hum Genet. 2017;100:267–80.CrossRef
17.
go back to reference Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073–81.CrossRef Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc. 2009;4:1073–81.CrossRef
18.
go back to reference Adzhubei I, Jordan DM, Sunyaev SR. Predicting functional effect of human missense mutations using PolyPhen-2. Current Protocols in Human Genetics. 2013;Chapter 7:Unit7.20. Adzhubei I, Jordan DM, Sunyaev SR. Predicting functional effect of human missense mutations using PolyPhen-2. Current Protocols in Human Genetics. 2013;Chapter 7:Unit7.20.
19.
go back to reference Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–24.CrossRef Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405–24.CrossRef
20.
go back to reference Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29:24.CrossRef Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. Integrative genomics viewer. Nat Biotechnol. 2011;29:24.CrossRef
21.
go back to reference Tjensvoll K, Lapin M, Buhl T, Oltedal S, Steen-Ottosen Berry K, Gilje B, et al. Clinical relevance of circulating KRAS mutated DNA in plasma from patients with advanced pancreatic cancer. Mol Oncol. 2016;10:635–43.CrossRef Tjensvoll K, Lapin M, Buhl T, Oltedal S, Steen-Ottosen Berry K, Gilje B, et al. Clinical relevance of circulating KRAS mutated DNA in plasma from patients with advanced pancreatic cancer. Mol Oncol. 2016;10:635–43.CrossRef
22.
go back to reference Gilje B, Heikkila R, Oltedal S, Tjensvoll K, Nordgard O. High-fidelity DNA polymerase enhances the sensitivity of a peptide nucleic acid clamp PCR assay for K-ras mutations. J Mol Diagn. 2008;10:325–31.CrossRef Gilje B, Heikkila R, Oltedal S, Tjensvoll K, Nordgard O. High-fidelity DNA polymerase enhances the sensitivity of a peptide nucleic acid clamp PCR assay for K-ras mutations. J Mol Diagn. 2008;10:325–31.CrossRef
23.
go back to reference Immervoll H, Hoem D, Kugarajh K, Steine SJ, Molven A. Molecular analysis of the EGFR-RAS-RAF pathway in pancreatic ductal adenocarcinomas: lack of mutations in the BRAF and EGFR genes. Virchows Arch. 2006;448:788–96.CrossRef Immervoll H, Hoem D, Kugarajh K, Steine SJ, Molven A. Molecular analysis of the EGFR-RAS-RAF pathway in pancreatic ductal adenocarcinomas: lack of mutations in the BRAF and EGFR genes. Virchows Arch. 2006;448:788–96.CrossRef
24.
go back to reference Immervoll H, Hoem D, Steffensen OJ, Miletic H, Molven A. Visualization of CD44 and CD133 in normal pancreas and pancreatic ductal adenocarcinomas: non-overlapping membrane expression in cell populations positive for both markers. J Histochem Cytochem. 2011;59:441–55.CrossRef Immervoll H, Hoem D, Steffensen OJ, Miletic H, Molven A. Visualization of CD44 and CD133 in normal pancreas and pancreatic ductal adenocarcinomas: non-overlapping membrane expression in cell populations positive for both markers. J Histochem Cytochem. 2011;59:441–55.CrossRef
25.
go back to reference El Jellas K, Hoem D, Hagen KG, Kalvenes MB, Aziz S, Steine SJ, et al. Associations between ABO blood groups and pancreatic ductal adenocarcinoma: influence on resection status and survival. Cancer Medicine. 2017;6:1531–40.CrossRef El Jellas K, Hoem D, Hagen KG, Kalvenes MB, Aziz S, Steine SJ, et al. Associations between ABO blood groups and pancreatic ductal adenocarcinoma: influence on resection status and survival. Cancer Medicine. 2017;6:1531–40.CrossRef
26.
go back to reference Nigro JM, Baker SJ, Preisinger AC, Jessup JM, Hostetter R, Cleary K, et al. Mutations in the p53 gene occur in diverse human tumour types. Nature. 1989;342:705–8.CrossRef Nigro JM, Baker SJ, Preisinger AC, Jessup JM, Hostetter R, Cleary K, et al. Mutations in the p53 gene occur in diverse human tumour types. Nature. 1989;342:705–8.CrossRef
27.
go back to reference Yamaguchi Y, Watanabe H, Yrdiran S, Ohtsubo K, Motoo Y, Okai T, et al. Detection of mutations of p53 tumor suppressor gene in pancreatic juice and its application to diagnosis of patients with pancreatic cancer: comparison with K-ras mutation. Clin Cancer Res. 1999;5:1147–53.PubMed Yamaguchi Y, Watanabe H, Yrdiran S, Ohtsubo K, Motoo Y, Okai T, et al. Detection of mutations of p53 tumor suppressor gene in pancreatic juice and its application to diagnosis of patients with pancreatic cancer: comparison with K-ras mutation. Clin Cancer Res. 1999;5:1147–53.PubMed
28.
go back to reference Sadakari Y, Kanda M, Maitani K, Borges M, Canto MI, Goggins M. Mutant KRAS and GNAS DNA concentrations in secretin-stimulated pancreatic fluid collected from the pancreatic duct and the duodenal lumen. Clin Transl Gastroenterol. 2014;5:e62.CrossRef Sadakari Y, Kanda M, Maitani K, Borges M, Canto MI, Goggins M. Mutant KRAS and GNAS DNA concentrations in secretin-stimulated pancreatic fluid collected from the pancreatic duct and the duodenal lumen. Clin Transl Gastroenterol. 2014;5:e62.CrossRef
29.
go back to reference Suenaga M, Dudley B, Karloski E, Borges M, Irene Canto M, Brand RE, et al. The effect of pancreatic juice collection time on the detection of KRAS mutations. Pancreas. 2018;47:35–9.CrossRef Suenaga M, Dudley B, Karloski E, Borges M, Irene Canto M, Brand RE, et al. The effect of pancreatic juice collection time on the detection of KRAS mutations. Pancreas. 2018;47:35–9.CrossRef
30.
go back to reference Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, et al. Extended RAS gene mutation testing in metastatic colorectal carcinoma to predict response to anti–epidermal growth factor receptor monoclonal antibody therapy: American Society of Clinical Oncology provisional clinical opinion update 2015. J Clin Oncol. 2016;34:179–85.CrossRef Allegra CJ, Rumble RB, Hamilton SR, Mangu PB, Roach N, Hantel A, et al. Extended RAS gene mutation testing in metastatic colorectal carcinoma to predict response to anti–epidermal growth factor receptor monoclonal antibody therapy: American Society of Clinical Oncology provisional clinical opinion update 2015. J Clin Oncol. 2016;34:179–85.CrossRef
31.
go back to reference Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.CrossRef Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, et al. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.CrossRef
32.
go back to reference Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.CrossRef Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.CrossRef
33.
go back to reference Andea A, Sarkar F, Adsay VN. Clinicopathological correlates of pancreatic intraepithelial neoplasia: a comparative analysis of 82 cases with and 152 cases without pancreatic ductal adenocarcinoma. Mod Pathol. 2003;16:996–1006.CrossRef Andea A, Sarkar F, Adsay VN. Clinicopathological correlates of pancreatic intraepithelial neoplasia: a comparative analysis of 82 cases with and 152 cases without pancreatic ductal adenocarcinoma. Mod Pathol. 2003;16:996–1006.CrossRef
34.
go back to reference Laghi L, Orbetegli O, Bianchi P, Zerbi A, Di Carlo V, Boland CR, et al. Common occurrence of multiple K-RAS mutations in pancreatic cancers with associated precursor lesions and in biliary cancers. Oncogene. 2002;21:4301–6.CrossRef Laghi L, Orbetegli O, Bianchi P, Zerbi A, Di Carlo V, Boland CR, et al. Common occurrence of multiple K-RAS mutations in pancreatic cancers with associated precursor lesions and in biliary cancers. Oncogene. 2002;21:4301–6.CrossRef
35.
go back to reference Makohon-Moore A, Iacobuzio-Donahue CA. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat Rev Cancer. 2016;16:553.CrossRef Makohon-Moore A, Iacobuzio-Donahue CA. Pancreatic cancer biology and genetics from an evolutionary perspective. Nat Rev Cancer. 2016;16:553.CrossRef
36.
go back to reference Schirmer M, Ijaz UZ, D'Amore R, Hall N, Sloan WT, Quince C. Insight into biases and sequencing errors for amplicon sequencing with the Illumina MiSeq platform. Nucleic Acids Res. 2015;43:e37.CrossRef Schirmer M, Ijaz UZ, D'Amore R, Hall N, Sloan WT, Quince C. Insight into biases and sequencing errors for amplicon sequencing with the Illumina MiSeq platform. Nucleic Acids Res. 2015;43:e37.CrossRef
37.
go back to reference Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci. 2011;108:9530–5.CrossRef Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci. 2011;108:9530–5.CrossRef
38.
go back to reference Erchinger F, Engjom T, Tjora E, Hoem D, Hausken T, Gilja OH, et al. Quantification of pancreatic function using a clinically feasible short endoscopic secretin test. Pancreas. 2013;42:1101–6.CrossRef Erchinger F, Engjom T, Tjora E, Hoem D, Hausken T, Gilja OH, et al. Quantification of pancreatic function using a clinically feasible short endoscopic secretin test. Pancreas. 2013;42:1101–6.CrossRef
39.
go back to reference Mueller S, Engleitner T, Maresch R, Zukowska M, Lange S, Kaltenbacher T, et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature. 2018;554:62–8.CrossRef Mueller S, Engleitner T, Maresch R, Zukowska M, Lange S, Kaltenbacher T, et al. Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature. 2018;554:62–8.CrossRef
40.
go back to reference Notta F, Chan-Seng-Yue M, Lemire M, Li Y, Wilson GW, Connor AA, et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature. 2016;538:378–82.CrossRef Notta F, Chan-Seng-Yue M, Lemire M, Li Y, Wilson GW, Connor AA, et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature. 2016;538:378–82.CrossRef
41.
go back to reference Yokoyama S, Kitamoto S, Higashi M, Goto Y, Hara T, Ikebe D, et al. Diagnosis of pancreatic neoplasms using a novel method of DNA methylation analysis of mucin expression in pancreatic juice. PLoS One. 2014;9:e93760.CrossRef Yokoyama S, Kitamoto S, Higashi M, Goto Y, Hara T, Ikebe D, et al. Diagnosis of pancreatic neoplasms using a novel method of DNA methylation analysis of mucin expression in pancreatic juice. PLoS One. 2014;9:e93760.CrossRef
42.
go back to reference Hata T, Ishida M, Motoi F, Yamaguchi T, Naitoh T, Katayose Y, et al. Telomerase activity in pancreatic juice differentiates pancreatic cancer from chronic pancreatitis: a meta-analysis. Pancreatology. 2016;16:372–81.CrossRef Hata T, Ishida M, Motoi F, Yamaguchi T, Naitoh T, Katayose Y, et al. Telomerase activity in pancreatic juice differentiates pancreatic cancer from chronic pancreatitis: a meta-analysis. Pancreatology. 2016;16:372–81.CrossRef
Metadata
Title
Mutation analysis by deep sequencing of pancreatic juice from patients with pancreatic ductal adenocarcinoma
Authors
Man Hung Choi
Eline Mejlænder-Andersen
Sophia Manueldas
Khadija El Jellas
Solrun J. Steine
Kjersti Tjensvoll
Hege Aase Sætran
Stian Knappskog
Dag Hoem
Oddmund Nordgård
Randi Hovland
Anders Molven
Publication date
01-12-2019
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2019
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-5195-7

Other articles of this Issue 1/2019

BMC Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine