Early and accurate detection of cholangiocarcinoma in patients with primary sclerosing cholangitis by methylation markers in bile

Hege Marie Vedeld, Marit M Grimsrud, Kim Andresen, Heidi D Pharo, Erik von Seth, Tom H Karlsen, Hilde Honne, Vemund Paulsen, Martti A Färkkilä, Annika Bergquist, Marine Jeanmougin, Lars Aabakken, Kirsten M Boberg, Trine Folseraas, Guro E Lind, Hege Marie Vedeld, Marit M Grimsrud, Kim Andresen, Heidi D Pharo, Erik von Seth, Tom H Karlsen, Hilde Honne, Vemund Paulsen, Martti A Färkkilä, Annika Bergquist, Marine Jeanmougin, Lars Aabakken, Kirsten M Boberg, Trine Folseraas, Guro E Lind

Abstract

Background and aims: Primary sclerosing cholangitis (PSC) is associated with increased risk of cholangiocarcinoma (CCA). Early and accurate CCA detection represents an unmet clinical need as the majority of patients with PSC are diagnosed at an advanced stage of malignancy. In the present study, we aimed at establishing robust DNA methylation biomarkers in bile for early and accurate diagnosis of CCA in PSC.

Approach and results: Droplet digital PCR (ddPCR) was used to analyze 344 bile samples from 273 patients with sporadic and PSC-associated CCA, PSC, and other nonmalignant liver diseases for promoter methylation of cysteine dioxygenase type 1, cannabinoid receptor interacting protein 1, septin 9, and vimentin. Receiver operating characteristic (ROC) curve analyses revealed high AUCs for all four markers (0.77-0.87) for CCA detection among patients with PSC. Including only samples from patients with PSC diagnosed with CCA ≤ 12 months following bile collection increased the accuracy for cancer detection, with a combined sensitivity of 100% (28/28) and a specificity of 90% (20/203). The specificity increased to 93% when only including patients with PSC with longtime follow-up (> 36 months) as controls, and remained high (83%) when only including patients with PSC and dysplasia as controls (n = 23). Importantly, the bile samples from the CCA-PSC ≤ 12 patients, all positive for the biomarkers, included both early-stage and late-stage CCA, different tumor growth patterns, anatomical locations, and carbohydrate antigen 19-9 levels.

Conclusions: Using highly sensitive ddPCR to analyze robust epigenetic biomarkers, CCA in PSC was accurately detected in bile, irrespective of clinical and molecular features, up to 12 months before CCA diagnosis. The findings suggest a potential for these biomarkers to complement current detection and screening methods for CCA in patients with PSC.

Conflict of interest statement

Dr. Bergquist received grants from Gilead.

© 2021 The Authors. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.

Figures

FIGURE 1
FIGURE 1
Flowchart of bile samples included in the study. Number of patients, followed by number of samples in parentheses. Samples are split based on main disease category, including (1) patients with underlying PSC (left panel) and (2) patients with non‐PSC‐related diseases (right panel). PSC samples (left panel) are divided into the following groups: (1) CCA‐PSC (n = 38, 42 samples), including CCA‐PSC ≤ 12 (n = 28, 31 samples; ≤ 12 months from bile sampling to CCA diagnosis) and CCA‐PSC > 12 (n = 10, 11 samples; > 12 months from bile sampling to CCA diagnosis), and (2) all PSC samples (without CCA, n = 205, 272 samples), including PSC‐dysplasia (n = 23, 24 samples; patients with PSC with evidence of biliary dysplasia based on assessment of biliary brush cytology specimens or histological assessment of explant liver ±2 months from bile collection), PSC‐control > 36 (n = 170, 226 samples; patients with PSC with no evidence of CCA or biliary dysplasia based on histological assessment of explanted liver at or after bile collection or > 36 months of follow‐up without established CCA or biliary dysplasia for nontransplanted patients), and PSC‐control < 36 (n = 12, 22 samples; patients with PSC with no evidence of CCA or biliary dysplasia after bile collection but only including nontransplanted patients with < 36 months of follow‐up). Non‐PSC samples (right panel) are divided into (1) CCA (n = 6, 6 samples, of which five were collected at the same time as a confirmed CCA diagnosis and one was collected 3 months prior to diagnosis) and (2) NM liver disease (n = 24, 24 samples; hereditary, idiopathic nonalcoholic fatty liver, biliary stone, and autoimmune liver disease other than PSC)
FIGURE 2
FIGURE 2
ROC curves, calculated AUCs, and sensitivity and specificity values for the four individual DNA methylation biomarkers in bile. (A) Samples from patients with CCA‐PSC (n = 38) versus all PSC (n = 205), (B) samples from patients with PSC diagnosed with CCA ≤ 12 months after bile sampling (CCA‐PSC ≤ 12, n = 28) versus PSC all (n = 205), (C) CCA‐PSC ≤ 12 (n = 28) versus PSC‐control > 36 (n = 170, including samples from patients with PSC showing no evidence of biliary dysplasia or CCA in explanted liver or with > 36 months of follow‐up), and (D) CCA‐PSC ≤ 12 (n = 28) versus PSC‐dysplasia (n = 23, including patients with PSC with evidence of biliary dysplasia based on assessment of biliary brush cytology specimens or histological assessment of explant liver ±2 months from bile collection)
FIGURE 3
FIGURE 3
Comparison of DNA methylation in bile with CA 19‐9 levels and biliary brush cytology. Red circle, sample scored as positive for methylation/cytology/CA 19‐9; white circle, sample scored as negative for methylation/cytology/CA 19‐9. The methylation panel was considered positive if one or more of the four biomarkers were methylated. Biliary brush cytology was scored positive if low/moderate‐grade or high‐grade dysplasia was identified and negative if only normal cells were identified. If more than one brush sample was taken, the case was scored based on the highest grade of dysplasia found. In cases where the sample was scored between two grades of dysplasia, the highest grade was used. For CA 19‐9 levels, 100 U/ml was used as the threshold: a CA 19‐9 level > 100 U/ml was considered positive, while a CA 19‐9 level

References

    1. Hirschfield GM, Karlsen TH, Lindor KD, Adams DH. Primary sclerosing cholangitis. Lancet. 2013;382:1587–99.
    1. Schrumpf E, Boberg KM, Karlsen TH. Primary sclerosing cholangitis—the Norwegian experience. Scand J Gastroenterol. 2015;50:781–96.
    1. Karlsen TH, Folseraas T, Thorburn D, Vesterhus M. Primary sclerosing cholangitis—a comprehensive review. J Hepatol. 2017;67:1298–323.
    1. Bergquist A, Ekbom A, Olsson R, Kornfeldt D, Lööf L, Danielsson Å, et al. Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. J Hepatol. 2002;36:321–7.
    1. Boonstra K, Weersma RK, van Erpecum KJ, Rauws EA, Spanier BWM, Poen AC, et al. Population‐based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis. Hepatology. 2013;58:2045–55.
    1. Tanaka A, Takamori Y, Toda G, Ohnishi S, Takikawa H. Outcome and prognostic factors of 391 Japanese patients with primary sclerosing cholangitis. Liver Int. 2008;28:983–9.
    1. Weismuller TJ, Trivedi PJ, Bergquist A, Imam M, Lenzen H, Ponsioen CY, et al. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology. 2017;152:1975–84.e8.
    1. Folseraas T, Boberg KM. Cancer risk and surveillance in primary sclerosing cholangitis. Clin Liver Dis. 2016;20:79–98.
    1. Razumilava N, Gores GJ. Surveillance for cholangiocarcinoma in patients with primary sclerosing cholangitis: effective and justified? Clin Liver Dis. 2016;8:43–7.
    1. Bowlus CL, Lim JK, Lindor KD. AGA clinical practice update on surveillance for hepatobiliary cancers in patients with primary sclerosing cholangitis: expert review. Clin Gastroenterol Hepatol. 2019;17:2416–22.
    1. Kang MJ, Jang J‐Y, Chang J, Shin YC, Lee D, Kim HB, et al. Actual long‐term survival outcome of 403 consecutive patients with hilar cholangiocarcinoma. World J Surg. 2016;40:2451–9.
    1. Azad AI, Rosen CB, Taner T, Heimbach JK, Gores GJ. Selected patients with unresectable perihilar cholangiocarcinoma (pCCA) derive long‐term benefit from liver transplantation. Cancers. 2020;12(11):3157.
    1. Vedeld HM, Goel A, Lind GE. Epigenetic biomarkers in gastrointestinal cancers: the current state and clinical perspectives. Semin Cancer Biol. 2018;51:36–49.
    1. Vedeld HM, Folseraas T, Lind GE. Detecting cholangiocarcinoma in patients with primary sclerosing cholangitis—the promise of DNA methylation and molecular biomarkers. JHEP Rep. 2020;2:100143.
    1. Andresen K, Boberg KM, Vedeld HM, Honne H, Jebsen P, Hektoen M, et al. Four DNA methylation biomarkers in biliary brush samples accurately identify the presence of cholangiocarcinoma. Hepatology. 2015;61:1651–9.
    1. Lazaridis KN, LaRusso NF. Primary sclerosing cholangitis. N Engl J Med. 2016;375:1161–70.
    1. Rizvi S, Khan SA, Hallemeier CL, Kelley RK, Gores GJ. Cholangiocarcinoma—evolving concepts and therapeutic strategies. Nat Rev Clin Oncol. 2018;15:95–111.
    1. Majeed A, Castedal M, Arnelo U, Söderdahl G, Bergquist A, Said K. Optimizing the detection of biliary dysplasia in primary sclerosing cholangitis before liver transplantation. Scand J Gastroenterol. 2018;53:56–63.
    1. Boberg KM, Jebsen P, Clausen OP, Foss A, Aabakken L, Schrumpf E. Diagnostic benefit of biliary brush cytology in cholangiocarcinoma in primary sclerosing cholangitis. J Hepatol. 2006;45:568–74.
    1. Lind GE, Danielsen SA, Ahlquist T, Merok MA, Andresen K, Skotheim RI, et al. Identification of an epigenetic biomarker panel with high sensitivity and specificity for colorectal cancer and adenomas. Mol Cancer. 2011;10:85.
    1. Pharo HD, Andresen K, Berg KCG, Lothe RA, Jeanmougin M, Lind GE. A robust internal control for high‐precision DNA methylation analyses by droplet digital PCR. Clin Epigenetics. 2018;10:24.
    1. Andresen K, Boberg K, Vedeld H, Honne H, Hektoen M, Wadsworth C, et al. Novel target genes and a valid biomarker panel identified for cholangiocarcinoma. Epigenetics. 2012;7:1249–57.
    1. Klump B, Hsieh CJ, Dette S, Holzmann K, Kiebetalich R, Jung M, et al. Promoter methylation of INK4a/ARF as detected in bile—significance for the differential diagnosis in biliary disease. Clin Cancer Res. 2003;9:1773–8.
    1. Shin S‐H, Lee K, Kim B‐H, Cho N‐Y, Jang J‐Y, Kim Y‐T, et al. Bile‐based detection of extrahepatic cholangiocarcinoma with quantitative DNA methylation markers and its high sensitivity. J Mol Diagn. 2012;14:256–63.
    1. Barnicle A, Seoighe C, Greally JM, Golden A, Egan LJ. Inflammation‐associated DNA methylation patterns in epithelium of ulcerative colitis. Epigenetics. 2017;12:591–606.
    1. Metzger J, Negm AA, Plentz RR, Weismüller TJ, Wedemeyer J, Karlsen TH, et al. Urine proteomic analysis differentiates cholangiocarcinoma from primary sclerosing cholangitis and other benign biliary disorders. Gut. 2013;62:122–30.
    1. Lankisch TO, Metzger J, Negm AA, Voβkuhl K, Schiffer E, Siwy J, et al. Bile proteomic profiles differentiate cholangiocarcinoma from primary sclerosing cholangitis and choledocholithiasis. Hepatology. 2011;53:875–84.
    1. Branchi V, Schaefer P, Semaan A, Kania A, Lingohr P, Kalff JC, et al. Promoter hypermethylation of SHOX2 and SEPT9 is a potential biomarker for minimally invasive diagnosis in adenocarcinomas of the biliary tract. Clin Epigenetics. 2016;8:133.
    1. Razumilava N, Gores GJ, Lindor KD. Cancer surveillance in patients with primary sclerosing cholangitis. Hepatology. 2011;54:1842–52.
    1. Eaton JE, Gossard AA, Talwalkar JA. Recall processes for biliary cytology in primary sclerosing cholangitis. Curr Opin Gastroenterol. 2014;30:287–94.
    1. Rizvi S, Eaton JE, Gores GJ. Primary sclerosing cholangitis as a premalignant biliary tract disease: surveillance and management. Clin Gastroenterol Hepatol. 2015;13:2152–65.
    1. Charatcharoenwitthaya P, Enders FB, Halling KC, Lindor KD. Utility of serum tumor markers, imaging, and biliary cytology for detecting cholangiocarcinoma in primary sclerosing cholangitis. Hepatology. 2008;48:1106–17.
    1. Boyd S, Mustonen H, Tenca A, Jokelainen K, Arola J, Färkkilä MA. Surveillance of primary sclerosing cholangitis with ERC and brush cytology: risk factors for cholangiocarcinoma. Scand J Gastroenterol. 2017;52:242–9.
    1. Boyd S, Tenca A, Jokelainen K, Mustonen H, Krogerus L, Arola J, et al. Screening primary sclerosing cholangitis and biliary dysplasia with endoscopic retrograde cholangiography and brush cytology: risk factors for biliary neoplasia. Endoscopy. 2016;48:432–9.
    1. Eaton JE, Welle CL, Bakhshi Z, Sheedy SP, Idilman IS, Gores GJ, et al. Early cholangiocarcinoma detection with magnetic resonance imaging versus ultrasound in primary sclerosing cholangitis. Hepatology. 2021;73:1868–81.
    1. Njei B, McCarty TR, Varadarajulu S, Navaneethan U. Systematic review with meta‐analysis: endoscopic retrograde cholangiopancreatography‐based modalities for the diagnosis of cholangiocarcinoma in primary sclerosing cholangitis. Aliment Pharmacol Ther. 2016;44:1139–51.
    1. Trikudanathan G, Navaneethan U, Njei B, Vargo JJ, Parsi MA. Diagnostic yield of bile duct brushings for cholangiocarcinoma in primary sclerosing cholangitis: a systematic review and meta‐analysis. Gastrointest Endosc. 2014;79:783–9.
    1. Harewood GC, Baron TH, Stadheim LM, Kipp BR, Sebo TJ, Salomao DR. Prospective, blinded assessment of factors influencing the accuracy of biliary cytology interpretation. Am J Gastroenterol. 2004;99:1464–9.
    1. Lewis JT, Talwalkar JA, Rosen CB, Smyrk TC, Abraham SC. Precancerous bile duct pathology in end‐stage primary sclerosing cholangitis, with and without cholangiocarcinoma. Am J Surg Pathol. 2010;34:27–34.
    1. Nakamoto S, Kumamoto Y, Igarashi K, Fujiyama Y, Nishizawa N, Ei S, et al. Methylated promoter DNA of CDO1 gene and preoperative serum CA19‐9 are prognostic biomarkers in primary extrahepatic cholangiocarcinoma. PLoS ONE. 2018;13:e0205864.
    1. Chen D, Wu H, Feng X, Chen Y, Lv Z, Kota VG, et al. DNA methylation of cannabinoid receptor interacting protein 1 promotes pathogenesis of intrahepatic cholangiocarcinoma through suppressing Parkin‐dependent pyruvate kinase M2 ubiquitination. Hepatology. 2021;73(5):1816–35.

Source: PubMed

3
Iratkozz fel