Skip to main content
Top
Published in: Journal of Gastroenterology 12/2020

01-12-2020 | Pancreatic Cancer | Original Article—Liver, Pancreas, and Biliary Tract

Digital PCR-based plasma cell-free DNA mutation analysis for early-stage pancreatic tumor diagnosis and surveillance

Authors: Tetsuhiro Okada, Yusuke Mizukami, Yusuke Ono, Hiroki Sato, Akihiro Hayashi, Hidemasa Kawabata, Kazuya Koizumi, Sakue Masuda, Shinichi Teshima, Kuniyuki Takahashi, Akio Katanuma, Yuko Omori, Hirotoshi Iwano, Masataka Yamada, Tomoki Yokochi, Shingo Asahara, Kazumichi Kawakubo, Masaki Kuwatani, Naoya Sakamoto, Katsuro Enomoto, Takuma Goto, Junpei Sasajima, Mikihiro Fujiya, Jun Ueda, Seiji Matsumoto, Kenzui Taniue, Ayumu Sugitani, Hidenori Karasaki, Toshikatsu Okumura

Published in: Journal of Gastroenterology | Issue 12/2020

Login to get access

Abstract

Background

Cell-free DNA (cfDNA) shed from tumors into the circulation offers a tool for cancer detection. Here, we evaluated the feasibility of cfDNA measurement and utility of digital PCR (dPCR)-based assays, which reduce subsampling error, for diagnosing pancreatic ductal adenocarcinoma (PDA) and surveillance of intraductal papillary mucinous neoplasm (IPMN).

Methods

We collected plasma from seven institutions for cfDNA measurements. Hot-spot mutations in KRAS and GNAS in the cfDNA from patients with PDA (n = 96), undergoing surveillance for IPMN (n = 112), and normal controls (n = 76) were evaluated using pre-amplification dPCR.

Results

Upon Qubit measurement and copy number assessment of hemoglobin-subunit (HBB) and mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1 (MT-ND1) in plasma cfDNA, HBB offered the best resolution between patients with PDA relative to healthy subjects [area under the curve (AUC) 0.862], whereas MT-ND1 revealed significant differences between IPMN and controls (AUC 0.851). DPCR utilizing pre-amplification cfDNA afforded accurate tumor-derived mutant KRAS detection in plasma in resectable PDA (AUC 0.861–0.876) and improved post-resection recurrence prediction [hazard ratio (HR) 3.179, 95% confidence interval (CI) 1.025–9.859] over that for the marker CA19-9 (HR 1.464; 95% CI 0.674–3.181). Capturing KRAS and GNAS could also provide genetic evidence in patients with IPMN-associated PDA and undergoing pancreatic surveillance.

Conclusions

Plasma cfDNA quantification by distinct measurements is useful to predict tumor burden. Through appropriate methods, dPCR-mediated mutation detection in patients with localized PDA and IPMN likely to progress to invasive carcinoma is feasible and complements conventional biomarkers.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kanda M, Matthaei H, Wu J, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142:730–9.CrossRef Kanda M, Matthaei H, Wu J, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology. 2012;142:730–9.CrossRef
2.
go back to reference Patra KC, Bardeesy N, Mizukami Y. Diversity of precursor lesions for pancreatic cancer: the genetics and biology of intraductal papillary mucinous neoplasm. Clin Transl Gastroenterol. 2017;8:e86.CrossRef Patra KC, Bardeesy N, Mizukami Y. Diversity of precursor lesions for pancreatic cancer: the genetics and biology of intraductal papillary mucinous neoplasm. Clin Transl Gastroenterol. 2017;8:e86.CrossRef
3.
go back to reference Gormally E, Vineis P, Matullo G, et al. TP53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: a prospective study. Cancer Res. 2006;66:6871–6.CrossRef Gormally E, Vineis P, Matullo G, et al. TP53 and KRAS2 mutations in plasma DNA of healthy subjects and subsequent cancer occurrence: a prospective study. Cancer Res. 2006;66:6871–6.CrossRef
4.
go back to reference Suenaga M, Yu J, Shindo K, 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, 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
5.
go back to reference Wu J, Matthaei H, Maitra A, et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med. 2011;3:92ra66.CrossRef Wu J, Matthaei H, Maitra A, et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med. 2011;3:92ra66.CrossRef
6.
go back to reference Furukawa T, Kuboki Y, Tanji E, et al. Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas. Sci Rep. 2011;1:161.CrossRef Furukawa T, Kuboki Y, Tanji E, et al. Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas. Sci Rep. 2011;1:161.CrossRef
7.
go back to reference Berger AW, Schwerdel D, Costa IG, et al. Detection of hot-spot mutations in circulating cell-free DNA from patients with intraductal papillary mucinous neoplasms of the pancreas. Gastroenterology. 2016;151:267–70.CrossRef Berger AW, Schwerdel D, Costa IG, et al. Detection of hot-spot mutations in circulating cell-free DNA from patients with intraductal papillary mucinous neoplasms of the pancreas. Gastroenterology. 2016;151:267–70.CrossRef
8.
go back to reference Wan JCM, Massie C, Garcia-Corbacho J, 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, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat Rev Cancer. 2017;17:223–38.CrossRef
9.
go back to reference Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.CrossRef Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra24.CrossRef
10.
go back to reference Takai E, Totoki Y, Nakamura H, et al. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci Rep. 2015;5:18425.CrossRef Takai E, Totoki Y, Nakamura H, et al. Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer. Sci Rep. 2015;5:18425.CrossRef
11.
go back to reference Hadano N, Murakami Y, Uemura K, et al. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br J Cancer. 2016;115:59–655.CrossRef Hadano N, Murakami Y, Uemura K, et al. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br J Cancer. 2016;115:59–655.CrossRef
12.
go back to reference Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–30.CrossRef Cohen JD, Li L, Wang Y, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–30.CrossRef
13.
go back to reference Kinde I, Wu J, Papadopoulos N, et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA. 2011;108:9530–5.CrossRef Kinde I, Wu J, Papadopoulos N, et al. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci USA. 2011;108:9530–5.CrossRef
14.
go back to reference Ono Y, Sugitani A, Karasaki H, et al. An improved digital polymerase chain reaction protocol to capture low-copy KRAS mutations in plasma cell-free DNA by resolving ‘subsampling’ issues. Mol Oncol. 2017;11:1448–58.CrossRef Ono Y, Sugitani A, Karasaki H, et al. An improved digital polymerase chain reaction protocol to capture low-copy KRAS mutations in plasma cell-free DNA by resolving ‘subsampling’ issues. Mol Oncol. 2017;11:1448–58.CrossRef
15.
go back to reference Rowlands V, Rutkowski AJ, Meuser E, et al. Optimisation of robust singleplex and multiplex droplet digital PCR assays for high confidence mutation detection in circulating tumour DNA. Sci Rep. 2019;9:12620.CrossRef Rowlands V, Rutkowski AJ, Meuser E, et al. Optimisation of robust singleplex and multiplex droplet digital PCR assays for high confidence mutation detection in circulating tumour DNA. Sci Rep. 2019;9:12620.CrossRef
16.
go back to reference Tanaka M, Fernandez-Del Castillo C, Kamisawa T, et al. Revisions of international consensus Fukuoka guidelines for the management of IPMN of the pancreas. Pancreatology. 2017;17:738–53.CrossRef Tanaka M, Fernandez-Del Castillo C, Kamisawa T, et al. Revisions of international consensus Fukuoka guidelines for the management of IPMN of the pancreas. Pancreatology. 2017;17:738–53.CrossRef
17.
go back to reference Del Chiaro M, Verbeke C, Salvia R, et al. European experts consensus statement on cystic tumours of the pancreas. Dig Liver Dis. 2013;45:703–11.CrossRef Del Chiaro M, Verbeke C, Salvia R, et al. European experts consensus statement on cystic tumours of the pancreas. Dig Liver Dis. 2013;45:703–11.CrossRef
18.
go back to reference Brierley JD, Gospodarowicz MK, Wittekind C. TNM classification of malignant tumours. 8th ed. Hoboken: Wiley-Blackwell; 2017. Brierley JD, Gospodarowicz MK, Wittekind C. TNM classification of malignant tumours. 8th ed. Hoboken: Wiley-Blackwell; 2017.
19.
go back to reference Omori Y, Ono Y, Tanino M, et al. Pathways of progression from intraductal papillary mucinous neoplasm to pancreatic ductal adenocarcinoma based on molecular features. Gastroenterology. 2019;156:647–81.CrossRef Omori Y, Ono Y, Tanino M, et al. Pathways of progression from intraductal papillary mucinous neoplasm to pancreatic ductal adenocarcinoma based on molecular features. Gastroenterology. 2019;156:647–81.CrossRef
20.
go back to reference Gemenetzis G, Bagante F, Griffin JF, et al. Neutrophil-to-lymphocyte ratio is a predictive marker for invasive malignancy in intraductal papillary mucinous neoplasms of the pancreas. Ann Surg. 2017;266:339–45.CrossRef Gemenetzis G, Bagante F, Griffin JF, et al. Neutrophil-to-lymphocyte ratio is a predictive marker for invasive malignancy in intraductal papillary mucinous neoplasms of the pancreas. Ann Surg. 2017;266:339–45.CrossRef
21.
go back to reference Kim J, Bamlet WR, Oberg AL, et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19–9 blood markers. Sci Transl Med. 2017;9:eaah5583.CrossRef Kim J, Bamlet WR, Oberg AL, et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19–9 blood markers. Sci Transl Med. 2017;9:eaah5583.CrossRef
22.
go back to reference Liu R, Chen X, Du Y, et al. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin Chem. 2012;58:610–8.CrossRef Liu R, Chen X, Du Y, et al. Serum microRNA expression profile as a biomarker in the diagnosis and prognosis of pancreatic cancer. Clin Chem. 2012;58:610–8.CrossRef
23.
go back to reference Madhavan B, Yue S, Galli U, et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer. 2015;136:2616–27.CrossRef Madhavan B, Yue S, Galli U, et al. Combined evaluation of a panel of protein and miRNA serum-exosome biomarkers for pancreatic cancer diagnosis increases sensitivity and specificity. Int J Cancer. 2015;136:2616–27.CrossRef
24.
go back to reference Luo G, Liu C, Guo M, et al. Potential biomarkers in Lewis negative patients with pancreatic cancer. Ann Surg. 2017;265:800–5.CrossRef Luo G, Liu C, Guo M, et al. Potential biomarkers in Lewis negative patients with pancreatic cancer. Ann Surg. 2017;265:800–5.CrossRef
25.
go back to reference Koldby KM, Mortensen MB, Detlefsen S, Pfeiffer P, Thomassen M, Kruse TA. Tumor-specific genetic aberrations in cell-free DNA of gastroesophageal cancer patients. J Gastroenterol. 2019;54:108–21.CrossRef Koldby KM, Mortensen MB, Detlefsen S, Pfeiffer P, Thomassen M, Kruse TA. Tumor-specific genetic aberrations in cell-free DNA of gastroesophageal cancer patients. J Gastroenterol. 2019;54:108–21.CrossRef
26.
go back to reference Sikora K, Bedin C, Vicentini C, et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int J Biol Mark. 2015;30:e136–e141141.CrossRef Sikora K, Bedin C, Vicentini C, et al. Evaluation of cell-free DNA as a biomarker for pancreatic malignancies. Int J Biol Mark. 2015;30:e136–e141141.CrossRef
27.
go back to reference Okada T, Iwano H, Ono Y, et al. Utility of "liquid biopsy" using pancreatic juice for early detection of pancreatic cancer. Endosc Int Open. 2018;6:E1454–E14611461.CrossRef Okada T, Iwano H, Ono Y, et al. Utility of "liquid biopsy" using pancreatic juice for early detection of pancreatic cancer. Endosc Int Open. 2018;6:E1454–E14611461.CrossRef
28.
go back to reference Jackson JB, Choi DS, Luketich JD, et al. Multiplex preamplification of serum DNA to facilitate reliable detection of extremely rare cancer mutations in circulating DNA by digital PCR. J Mol Diagn. 2016;18:235–43.CrossRef Jackson JB, Choi DS, Luketich JD, et al. Multiplex preamplification of serum DNA to facilitate reliable detection of extremely rare cancer mutations in circulating DNA by digital PCR. J Mol Diagn. 2016;18:235–43.CrossRef
29.
go back to reference Schmidt B, Reinicke D, Reindl I, et al. Liquid biopsy—performance of the PAXgene(R) blood ccfDNA tubes for the isolation and characterization of cell-free plasma DNA from tumor patients. Clin Chim Acta. 2017;469:94–8.CrossRef Schmidt B, Reinicke D, Reindl I, et al. Liquid biopsy—performance of the PAXgene(R) blood ccfDNA tubes for the isolation and characterization of cell-free plasma DNA from tumor patients. Clin Chim Acta. 2017;469:94–8.CrossRef
Metadata
Title
Digital PCR-based plasma cell-free DNA mutation analysis for early-stage pancreatic tumor diagnosis and surveillance
Authors
Tetsuhiro Okada
Yusuke Mizukami
Yusuke Ono
Hiroki Sato
Akihiro Hayashi
Hidemasa Kawabata
Kazuya Koizumi
Sakue Masuda
Shinichi Teshima
Kuniyuki Takahashi
Akio Katanuma
Yuko Omori
Hirotoshi Iwano
Masataka Yamada
Tomoki Yokochi
Shingo Asahara
Kazumichi Kawakubo
Masaki Kuwatani
Naoya Sakamoto
Katsuro Enomoto
Takuma Goto
Junpei Sasajima
Mikihiro Fujiya
Jun Ueda
Seiji Matsumoto
Kenzui Taniue
Ayumu Sugitani
Hidenori Karasaki
Toshikatsu Okumura
Publication date
01-12-2020
Publisher
Springer Singapore
Published in
Journal of Gastroenterology / Issue 12/2020
Print ISSN: 0944-1174
Electronic ISSN: 1435-5922
DOI
https://doi.org/10.1007/s00535-020-01724-5

Other articles of this Issue 12/2020

Journal of Gastroenterology 12/2020 Go to the issue