Cell-free DNA promoter hypermethylation in plasma as a diagnostic marker for pancreatic adenocarcinoma

Stine Dam Henriksen, Poul Henning Madsen, Anders Christian Larsen, Martin Berg Johansen, Asbjørn Mohr Drewes, Inge Søkilde Pedersen, Henrik Krarup, Ole Thorlacius-Ussing, Stine Dam Henriksen, Poul Henning Madsen, Anders Christian Larsen, Martin Berg Johansen, Asbjørn Mohr Drewes, Inge Søkilde Pedersen, Henrik Krarup, Ole Thorlacius-Ussing

Abstract

Background: Pancreatic cancer has a 5-year survival rate of only 5-7%. Difficulties in detecting pancreatic cancer at early stages results in the high mortality and substantiates the need for additional diagnostic tools. Surgery is the only curative treatment and unfortunately only possible in localized tumours. A diagnostic biomarker for pancreatic cancer will have a major impact on patient survival by facilitating early detection and the possibility for curative treatment. DNA promoter hypermethylation is a mechanism of early carcinogenesis, which can cause inactivation of tumour suppressor genes. The aim of this study was to examine promoter hypermethylation in a panel of selected genes from cell-free DNA, as a diagnostic marker for pancreatic adenocarcinoma.

Methods: Patients with suspected or biopsy-verified pancreatic cancer were included prospectively and consecutively. Patients with chronic/acute pancreatitis were included as additional benign control groups. Based on an optimized accelerated bisulfite treatment protocol, methylation-specific PCR of a 28 gene panel was performed on plasma samples. A diagnostic prediction model was developed by multivariable logistic regression analysis using backward stepwise elimination.

Results: Patients with pancreatic adenocarcinoma (n = 95), chronic pancreatitis (n = 97) and acute pancreatitis (n = 59) and patients screened, but negative for pancreatic adenocarcinoma (n = 27), were included. The difference in mean number of methylated genes in the cancer group (8.41 (95% CI 7.62-9.20)) vs the total control group (4.74 (95% CI 4.40-5.08)) was highly significant (p < 0.001). A diagnostic prediction model (age >65, BMP3, RASSF1A, BNC1, MESTv2, TFPI2, APC, SFRP1 and SFRP2) had an area under the curve of 0.86 (sensitivity 76%, specificity 83%). The model performance was independent of cancer stage.

Conclusions: Cell-free DNA promoter hypermethylation has the potential to be a diagnostic marker for pancreatic adenocarcinoma and differentiate between malignant and benign pancreatic disease. This study brings us closer to a clinical useful diagnostic marker for pancreatic cancer, which is urgently needed. External validation is, however, required before the test can be applied in the clinic.

Trial registration: ClinicalTrials.gov, NCT02079363.

Keywords: Cell-free DNA; Diagnostic biomarker; Epigenetic; Methylation; Pancreatic adenocarcinoma; Pancreatic cancer; Pancreatitis.

Figures

Fig. 1
Fig. 1
Flow diagram of patients included in the study. a Inclusion of patients with pancreatic adenocarcinoma. b Inclusion of patients with chronic pancreatitis. c Inclusion of patients with acute pancreatitis
Fig. 2
Fig. 2
Stepwise selection of genes for the pancreatic cancer diagnostic prediction model. Stepwise selection of genes with the corresponding p value and the area under the receiver operating characteristic curve (AUC). Model 13 was determined as the model with the best performance
Fig. 3
Fig. 3
Performance of model 13. a Stage I, II, III and IV pancreatic cancer. Model 13 (age >65, BMP3, RASSF1A, BNC1, MESTv2, TFPI2, APC, SFRP1, SFRP2). AUC = 0.86 (probability cut point of 0.50; sensitivity 76% and specificity 83%).b Stage I and II pancreatic cancer. Model 13 (age >65, BMP3, RASSF1A, BNC1, MESTv2, TFPI2, APC, SFRP1, SFRP2). AUC = 0.86 (probability cut point of 0.50; sensitivity 73% and specificity 83%)

References

    1. American Cancer Society: Pancreatic cancer survival rates, by stage [Online material]. Accessed Nov. 2016.
    1. Jemal A, Bray F, Ferlay J, Al E. Global Cancer Statistics. CA Cancer J CLIN. 2011;61:69–90.
    1. Michl P, Gress TM. Current concepts and novel targets in advanced pancreatic cancer. Gut. 2012;62(figure 1):317–26.
    1. Park JW, Baek IH, Kim YT. Preliminary study analyzing the methylated genes in the plasma of patients with pancreatic cancer. Scand J Surg. 2012;101:38–44. doi: 10.1177/145749691210100108.
    1. Haab BB, Huang Y, Balasenthil S, Partyka K, Tang H, Anderson M, Allen P, Sasson A, Zeh H, Kaul K, Kletter D, Ge S, Bern M, Kwon R, Blasutig I, Srivastava S, Frazier ML, Sen S, Hollingsworth MA, Rinaudo JA, Killary AM, Brand RE. Definitive characterization of CA 19-9 in resectable pancreatic cancer using a reference set of serum and plasma specimens. PLoS One. 2015;10:e0139049. doi: 10.1371/journal.pone.0139049.
    1. Hartwig W, Strobel O, Hinz U, Fritz S, Hackert T, Roth C, Büchler MW, Werner J. CA19-9 in potentially resectable pancreatic cancer: perspective to adjust surgical and perioperative therapy. Ann Surg Oncol. 2013;7:2188–96. doi: 10.1245/s10434-012-2809-1.
    1. Kim J-E, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004;19:182–6. doi: 10.1111/j.1440-1746.2004.03219.x.
    1. Delpu Y, Hanoun N, Lulka H, Sicard F, Selves J, Buscail L, Torrisani J, Cordelier P. Genetic and epigenetic alterations in pancreatic carcinogenesis. Curr Genomics. 2011;12:15–24. doi: 10.2174/138920211794520132.
    1. Lomberk GA. Epigenetic silencing of tumor suppressor genes in pancreatic cancer. J Gastrointest Cancer. 2011;42:93–9. doi: 10.1007/s12029-011-9256-2.
    1. Lomberk G, Mathison AJ, Grzenda A, Urrutia R. The sunset of somatic genetics and the dawn of epigenetics: a new frontier in pancreatic cancer research. Curr Opin Gastroenterol. 2008;24:597–602. doi: 10.1097/MOG.0b013e32830b111d.
    1. Mulero-Navarro S, Esteller M. Epigenetic biomarkers for human cancer: the time is now. Crit Rev Oncol Hematol. 2008;68:1–11. doi: 10.1016/j.critrevonc.2008.03.001.
    1. Costa FF. Epigenomics in cancer management. Cancer Manag Res. 2010;2:255–65. doi: 10.2147/CMAR.S7280.
    1. Sebova K, Fridrichova I. Epigenetic tools in potential anticancer therapy. Anticancer Drugs. 2010;21:565–77. doi: 10.1097/CAD.0b013e32833a4352.
    1. Jiao L, Zhu J, Hassan MM, Evans DB, Abbruzzese JL, Li D. K-ras mutation and p16 and preproenkephalin promoter hypermethylation in plasma DNA of pancreatic cancer patients: in relation to cigarette smoking. Pancreas. 2007;34:55–62. doi: 10.1097/01.mpa.0000246665.68869.d4.
    1. Esteller M, Rosell R, Sidransky D, Baylin SB. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999;59:67–70.
    1. Henriksen SD, Madsen PH, Krarup H, Thorlacius-Ussing O. DNA hypermethylation as a blood-based marker for pancreatic cancer: a literature review. Pancreas. 2015;44:1036–45. doi: 10.1097/MPA.0000000000000487.
    1. Yi JM, Guzzetta A a, Bailey VJ, Downing SR, Van Neste L, Chiappinelli KB, Keeley BP, Stark A, Herrera A, Wolfgang C, Pappou EP, Iacobuzio-Donahue C a, Goggins MG, Herman JG, Wang T-H, Baylin SB, Ahuja N. Novel methylation biomarker panel for the early detection of pancreatic cancer. Clin Cancer Res. 2013;19:6544–55. doi: 10.1158/1078-0432.CCR-12-3224.
    1. Larsen AC, Dabrowski T, Frøkjær JB, Fisker RV, Iyer VV, Møller BK, Kristensen SR, Thorlacius-Ussing O. Prevalence of venous thromboembolism at diagnosis of upper gastrointestinal cancer. Br J Surg. 2014;101:246–53. doi: 10.1002/bjs.9353.
    1. Jespersen J, Bertina R, Haverkate F. Laboratory techniques in thrombosis—a manual: second revised edition of the ECAT assay procedures. The Netherlands: Kluwer Academic Publishers, Dordrecht; 1999.
    1. Pedersen IS, Krarup HB, Thorlacius-Ussing O, Madsen PH. High recovery of cell-free methylated DNA based on a rapid bisulfite-treatment protocol. BMC Mol Biol. 2012;13:12. doi: 10.1186/1471-2199-13-12.
    1. Li L-C, Dahiya R. MethPrimer: designing primers for methylation PCRs. Bioinformatics. 2002;18:1427–31. doi: 10.1093/bioinformatics/18.11.1427.
    1. Mouliere F, Rosenfeld N. Circulating tumor-derived DNA is shorter than somatic DNA in plasma. Proc Natl Acad Sci. 2015;112:3178–9. doi: 10.1073/pnas.1501321112.
    1. Smith GCS, Seaman SR, Wood AM, Royston P, White IR. Correcting for optimistic prediction in small data sets. Am J Epidemiol. 2014;180:318–24. doi: 10.1093/aje/kwu140.
    1. Zhao Y, Sun J, Zhang H, Guo S, Gu J, Wang W, Tang N, Zhou X, Yu J. High-frequency aberrantly methylated targets in pancreatic adenocarcinoma identified via global DNA methylation analysis using methylCap-seq. Clin Epigenetics. 2014;6:18. doi: 10.1186/1868-7083-6-18.
    1. Vincent A, Omura N, Hong SM-M, Jaffe A, Eshleman J, Goggins M. Genome-wide analysis of promoter methylation associated with gene expression profile in pancreatic adenocarcinoma. Clin Cancer Res. 2011;17:4341–54. doi: 10.1158/1078-0432.CCR-10-3431.
    1. Park JK, Ryu JK, Yoon WJ, Lee SH, Lee GY, Jeong KS-S, Kim YT-T, Yoon YB. The role of quantitative NPTX2 hypermethylation as a novel serum diagnostic marker in pancreatic cancer. Pancreas. 2012;41:95–101. doi: 10.1097/MPA.0b013e318221c903.
    1. Kawasaki H, Igawa E, Kohosozawa R, Kobayashi M, Nishiko R, Abe H. Detection of aberrant methylation of tumor suppressor genes in plasma from cancer patients. Pers Med Universe. 2013;2:20–4. doi: 10.1016/j.pmu.2013.04.007.
    1. Liggett T, Melnikov A, Yi Q-LL, Replogle C, Brand R, Kaul K, Talamonti M, Abrams RA, Levenson V. Differential methylation of cell-free circulating DNA among patients with pancreatic cancer versus chronic pancreatitis. Cancer. 2010;116:1674–80. doi: 10.1002/cncr.24893.
    1. Peng DF-F, Kanai Y, Sawada M, Ushijima S, Hiraoka N, Kitazawa S, Hirohashi S. DNA methylation of multiple tumor-related genes in association with overexpression of DNA methyltransferase 1 (DNMT1) during multistage carcinogenesis of the pancreas. Carcinogenesis. 2006;27:1160–8. doi: 10.1093/carcin/bgi361.
    1. Li G, Ji Y, Liu C, Li J, Zhou Y. Reduced levels of p15INK4b, p16INK4a, p21cip1 and p27kip1 in pancreatic carcinoma. Mol Med Rep. 2012;5:1106–10.
    1. Li M, Zhao ZW. Clinical implications of mismatched repair gene promoter methylation in pancreatic cancer. Med Oncol. 2012;29:970–6. doi: 10.1007/s12032-011-9968-y.
    1. Ueki T, Toyota M, Sohn T, Yeo CJ, Issa JP, Hruban RH, Goggins M. Hypermethylation of multiple genes in pancreatic adenocarcinoma. Cancer Res. 2000;60:1835–9.
    1. Zhao G, Qin Q, Zhang J, Liu Y, Deng S, Liu L, Wang B, Tian K, Wang C. Hypermethylation of HIC1 promoter and aberrant expression of HIC1/SIRT1 might contribute to the carcinogenesis of pancreatic cancer. Ann Surg Oncol. 2013;20:S301-S311.
    1. Ballehaninna UK, Chamberlain RS. Serum CA 19-9 as a biomarker for pancreatic cancer—a comprehensive review. Indian J Surg Oncol. 2011;2:88–100. doi: 10.1007/s13193-011-0042-1.
    1. Sinsheimer JS, Bocklandt S, Lin W, Sehl ME, Sa FJ, Vilain E. Epigenetic predictor of age. PLoS One. 2011;6:1–6.
    1. Kurdyukov S, Bullock M. DNA methylation analysis: choosing the right method. Biology (Basel) 2016;5:3.

Source: PubMed

3
Abonneren