Promoter methylation of ADAMTS1 and BNC1 as potential biomarkers for early detection of pancreatic cancer in blood

Maryam A L Eissa, Lane Lerner, Eihab Abdelfatah, Nakul Shankar, Joseph K Canner, Nesrin M Hasan, Vesal Yaghoobi, Barry Huang, Zachary Kerner, Felipe Takaesu, Christopher Wolfgang, Ruby Kwak, Michael Ruiz, Matthew Tam, Thomas R Pisanic 2nd, Christine A Iacobuzio-Donahue, Ralph H Hruban, Jin He, Tza-Huei Wang, Laura D Wood, Anup Sharma, Nita Ahuja, Maryam A L Eissa, Lane Lerner, Eihab Abdelfatah, Nakul Shankar, Joseph K Canner, Nesrin M Hasan, Vesal Yaghoobi, Barry Huang, Zachary Kerner, Felipe Takaesu, Christopher Wolfgang, Ruby Kwak, Michael Ruiz, Matthew Tam, Thomas R Pisanic 2nd, Christine A Iacobuzio-Donahue, Ralph H Hruban, Jin He, Tza-Huei Wang, Laura D Wood, Anup Sharma, Nita Ahuja

Abstract

Background: Despite improvements in cancer management, most pancreatic cancers are still diagnosed at an advanced stage. We have recently identified promoter DNA methylation of the genes ADAMTS1 and BNC1 as potential blood biomarkers of pancreas cancer. In this study, we validate this biomarker panel in peripheral cell-free tumor DNA of patients with pancreatic cancer.

Results: Sensitivity and specificity for each gene are as follows: ADAMTS1 87.2% and 95.8% (AUC = 0.91; 95% CI 0.71-0.86) and BNC1 64.1% and 93.7% (AUC = 0.79; 95% CI 0.63-0.78). When using methylation of either gene as a combination panel, sensitivity increases to 97.3% and specificity to 91.6% (AUC = 0.95; 95% CI 0.77-0.90). Adding pre-operative CA 19-9 values to the combined two-gene methylation panel did not improve sensitivity. Methylation of ADAMTS1 was found to be positive in 87.5% (7/8) of stage I, 77.8% (7/9) of stage IIA, and 90% (18/20) of stage IIB disease. Similarly, BNC1 was positive in 62.5% (5/8) of stage I patients, 55.6% (5/9) of stage IIA, and 65% (13/20) of patients with stage IIB disease. The two-gene panel (ADAMTS1 and/or BNC1) was positive in 100% (8/8) of stage I, 88.9% (8/9) of stage IIA, and 100% (20/20) of stage IIB disease. The sensitivity and specificity of the two-gene panel for localized pancreatic cancer (stages I and II), where the cancer is eligible for surgical resection with curative potential, was 94.8% and 91.6% respectively. Additionally, the two-gene panel exhibited an AUC of 0.95 (95% CI 0.90-0.98) compared to 57.1% for CA 19-9 alone.

Conclusion: The methylation status of ADAMTS1 and BNC1 in cfDNA shows promise for detecting pancreatic cancer during the early stages when curative resection of the tumor is still possible. This minimally invasive blood-based biomarker panel could be used as a promising tool for diagnosis and screening in a select subset of high-risk populations.

Keywords: ADAMTS1; BNC1; Biomarker; Cell-free DNA; MOB; Methylation; Pancreatic cancer.

Conflict of interest statement

Ethics approval and consent to participate

The Institutional Review Boards of Johns Hopkins University approved the present study. In addition, blood samples were collected and analyzed as per institutional IRB protocol (NA00033085).

Consent for publication

Not applicable

Competing interests

NA has received grant funding from Cepheid and Astex and has served as a consultant to Ethicon. NA has licensed methylation Biomarkers (BNC1 and ADAMTS1) to Cepheid Inc. All other authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Methylation of ADAMTS1 and BNC1 genes in control and PDAC cases
Fig. 2
Fig. 2
Sensitivity and specificity of both genes. ROC curves for various genes. (a, b) ROC curves are represented for individual genes (ADAMTS1 and BNC1) and c combined methylation status of the genes (ADAMTS1 + BNC1) from the plasma samples

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.
    1. Bosetti C, et al. Pancreatic cancer: overview of descriptive epidemiology. Mol Carcinog. 2012;51(1):3–13.
    1. Worni M, et al. Modest improvement in overall survival for patients with metastatic pancreatic cancer: a trend analysis using the surveillance, epidemiology, and end results registry from 1988 to 2008. Pancreas. 2013;42(7):1157–1163.
    1. Karmazanovsky G, et al. Pancreatic head cancer: accuracy of CT in determination of resectability. Abdom Imaging. 2005;30(4):488–500.
    1. Garrido-Laguna I, Hidalgo M. Pancreatic cancer: from state-of-the-art treatments to promising novel therapies. Nat Rev Clin Oncol. 2015;12(6):319–334.
    1. Cancer Stat Facts: Pancreatic Cancer. Pancreatic Cancer - Cancer Stat Facts. 2018. .
    1. Ahuja N, Sharma AR, Baylin SB. Epigenetic therapeutics: a new weapon in the war against cancer. Annu Rev Med. 2016;67:73–89.
    1. Esteller M, et al. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Res. 1999;59(1):67–70.
    1. Chan TA, Baylin SB. Epigenetic biomarkers. Curr Top Microbiol Immunol. 2012;355:189–216.
    1. Hong SM, et al. Multiple genes are hypermethylated in intraductal papillary mucinous neoplasms of the pancreas. Mod Pathol. 2008;21(12):1499–1507.
    1. Hong SM, et al. Genome-wide CpG island profiling of intraductal papillary mucinous neoplasms of the pancreas. Clin Cancer Res. 2012;18(3):700–712.
    1. Sato N, et al. CpG island methylation profile of pancreatic intraepithelial neoplasia. Mod Pathol. 2008;21(3):238–244.
    1. Schuebel KE, et al. Comparing the DNA hypermethylome with gene mutations in human colorectal cancer. PLoS Genet. 2007;3(9):1709–1723.
    1. Jeschke J, et al. Biomarkers for detection and prognosis of breast cancer identified by a functional hypermethylome screen. Epigenetics. 2012;7(7):701–709.
    1. Yi JM, et al. Novel methylation biomarker panel for the early detection of pancreatic cancer. Clin Cancer Res. 2013;19(23):6544–6555.
    1. Hulbert A, et al. Early detection of lung cancer using DNA promoter hypermethylation in plasma and sputum. Clin Cancer Res. 2017;23(8):1998–2005.
    1. Matsubayashi H, et al. DNA methylation alterations in the pancreatic juice of patients with suspected pancreatic disease. Cancer Res. 2006;66(2):1208–1217.
    1. Kanda M, et al. Mutant GNAS detected in duodenal collections of secretin-stimulated pancreatic juice indicates the presence or emergence of pancreatic cysts. Gut. 2013;62(7):1024–1033.
    1. Frebourg T, et al. The evaluation of CA 19-9 antigen level in the early detection of pancreatic cancer. A prospective study of 866 patients. Cancer. 1988;62(11):2287–2290.
    1. Goonetilleke KS, et al. Diagnostic and prognostic value of plasma tumor M2 pyruvate kinase in periampullary cancer: evidence for a novel biological marker of adverse prognosis. Pancreas. 2007;34(3):318–324.
    1. Kaur S, et al. Early diagnosis of pancreatic cancer: challenges and new developments. Biomark Med. 2012;6(5):597–612.
    1. Steinberg W. The clinical utility of the CA 19-9 tumor-associated antigen. Am J Gastroenterol. 1990;85(4):350–355.
    1. Leon SA, et al. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 1977;37(3):646–650.
    1. Gahan PB, Swaminathan R. Circulating nucleic acids in plasma and serum. Recent developments. Ann N Y Acad Sci. 2008;1137:1–6.
    1. Karampini E, McCaughan F. Circulating DNA in solid organ cancers-analysis and clinical application. QJM. Int J Med. 2016;109(4):223–7.
    1. Cohen JD, et al. Combined circulating tumor DNA and protein biomarker-based liquid biopsy for the earlier detection of pancreatic cancers. Proc Natl Acad Sci U S A. 2017;114(38):10202–10207.
    1. Cohen JD, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359(6378):926–930.
    1. Guzzetta AA, et al. The promise of methylation on beads for cancer detection and treatment. Expert Rev Mol Diagn. 2014;14(7):845–852.
    1. Keeley B, et al. Extraction and processing of circulating DNA from large sample volumes using methylation on beads for the detection of rare epigenetic events. Clin Chim Acta. 2013;425:169–175.
    1. Pandol S, et al. Epidemiology, risk factors, and the promotion of pancreatic cancer: role of the stellate cell. J Gastroenterol Hepatol. 2012;27(Suppl 2):127–134.
    1. Yeo TP. Demographics, epidemiology, and inheritance of pancreatic ductal adenocarcinoma. Semin Oncol. 2015;42(1):8–18.
    1. Midha S, Chawla S, Garg PK. Modifiable and non-modifiable risk factors for pancreatic cancer: a review. Cancer Lett. 2016;381(1):269–277.
    1. A stool DNA test (Cologuard) for colorectal cancer screening. Jama. 2014;312(23):2566.
    1. Cai L, Hood S, Kallam E, Overman D, Barker K, et al. Epi proColon®: Use of a non-invasive SEPT9 gene methylation blood test for colorectal cancer screening: A national laboratory experience. J Clin Epigenet. 2018;4:7. 10.21767/2472-1158.100092.
    1. de Vos T, et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem. 2009;55(7):1337–1346.
    1. Warren JD, et al. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011;9:133.
    1. Rakovitch E, et al. Multigene expression assay and benefit of radiotherapy after breast conservation in ductal carcinoma in situ. J Natl Cancer Inst. 2017;109(4):djw256.
    1. Renfro LA, et al. Prospective evaluation of a 12-gene assay on patient treatment decisions and physician confidence in mismatch repair proficient stage IIA colon cancer. Clin Colorectal Cancer. 2017;16(1):23–30.
    1. Van Den Eeden SK, et al. A biopsy-based 17-gene genomic prostate score as a predictor of metastases and prostate cancer death in surgically treated men with clinically localized disease. Eur Urol. 2018;73(1):129–138.
    1. Esteller M, et al. A gene hypermethylation profile of human cancer. Cancer Res. 2001;61(8):3225–3229.
    1. Tsai HC, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res. 2011;21(3):502–517.
    1. Howlader N, Noone AM, Krapcho M, Miller D, Bishop K, Kosary CL, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA. SEER Cancer Statistics Review, 1975-2014. Bethesda: National Cancer Institute; 2017.
    1. Henschke CI, et al. Survival of patients with stage I lung cancer detected on CT screening. N Engl J Med. 2006;355(17):1763–1771.
    1. American Cancer Society . Colorectal Cancer Facts & Figures 2017-2019. Atlanta: American Cancer Society; 2017. p. 2017.
    1. Egawa S, et al. Clinicopathological aspects of small pancreatic cancer. Pancreas. 2004;28(3):235–240.
    1. Hur C, et al. Early pancreatic ductal adenocarcinoma survival is dependent on size: positive implications for future targeted screening. Pancreas. 2016;45(7):1062–6.
    1. Marchegiani G, et al. Does size matter in pancreatic cancer?: reappraisal of tumour dimension as a predictor of outcome beyond the TNM. Ann Surg. 2017;266(1):142–148.
    1. Lennon AM, et al. The early detection of pancreatic cancer: what will it take to diagnose and treat curable pancreatic neoplasia? Cancer Res. 2014;74(13):3381–3389.
    1. Wagner M, et al. Curative resection is the single most important factor determining outcome in patients with pancreatic adenocarcinoma. Br J Surg. 2004;91(5):586–594.
    1. Griffin JF, Poruk KE, Wolfgang CL. Pancreatic cancer surgery: past, present, and future. Chin J Cancer Res. 2015;27(4):332–348.
    1. Rulyak SJ, et al. Cost-effectiveness of pancreatic cancer screening in familial pancreatic cancer kindreds. Gastrointest Endosc. 2003;57(1):23–29.
    1. Shin EJ, Canto MI. Pancreatic cancer screening. Gastroenterol Clin N Am. 2012;41(1):143–157.
    1. Verna EC, et al. Pancreatic cancer screening in a prospective cohort of high-risk patients: a comprehensive strategy of imaging and genetics. Clin Cancer Res. 2010;16(20):5028–5037.
    1. Li D, et al. Pancreatic cancer. Lancet. 2004;363(9414):1049–1057.
    1. Petersen GM, et al. Pancreatic cancer genetic epidemiology consortium. Cancer Epidemiol Biomarkers Prev. 2006;15(4):704–710.
    1. Rustgi AK. Familial pancreatic cancer: genetic advances. Genes Dev. 2014;28(1):1–7.
    1. Yeo TP, et al. Pancreatic cancer. Curr Probl Cancer. 2002;26(4):176–275.
    1. Humphris JL, et al. Clinical and pathologic features of familial pancreatic cancer. Cancer. 2014;120(23):3669–3675.
    1. Petersen GM. Familial pancreatic adenocarcinoma. Hematol Oncol Clin North Am. 2015;29(4):641–653.
    1. Canto MI, et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut. 2013;62(3):339–347.
    1. Aberle DR, et al. Reduced lung-cancer mortality with low-dose computed tomographic screening. N Engl J Med. 2011;365(5):395–409.
    1. Anderson MA, et al. Alcohol and tobacco lower the age of presentation in sporadic pancreatic cancer in a dose-dependent manner: a multicenter study. Am J Gastroenterol. 2012;107(11):1730–1739.
    1. Pang T, et al. A novel scoring system to analyze combined effect of lifestyle factors on pancreatic cancer risk: a retrospective case-control study. Sci Rep. 2017;7(1):13657.
    1. Melo SA, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177–182.
    1. Babic A, Wolpin BM. Circulating exosomes in pancreatic cancer: will they succeed on the long, littered road to early detection marker? Clin Chem. 2016;62(2):307–9.
    1. Bailey VJ, et al. Single-tube analysis of DNA methylation with silica superparamagnetic beads. Clin Chem. 2010;56(6):1022–1025.
    1. Bailey VJ, et al. MS-qFRET: a quantum dot-based method for analysis of DNA methylation. Genome Res. 2009;19(8):1455–1461.
    1. Bustin SA, et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 2009;55(4):611–622.

Source: PubMed

3
구독하다