Integrative analysis reveals early and distinct genetic and epigenetic changes in intraductal papillary and tubulopapillary cholangiocarcinogenesis

Benjamin Goeppert, Damian Stichel, Reka Toth, Sarah Fritzsche, Moritz Anton Loeffler, Anna Melissa Schlitter, Olaf Neumann, Yassen Assenov, Monika Nadja Vogel, Arianeb Mehrabi, Katrin Hoffmann, Bruno Köhler, Christoph Springfeld, Dieter Weichenhan, Christoph Plass, Irene Esposito, Peter Schirmacher, Andreas von Deimling, Stephanie Roessler, Benjamin Goeppert, Damian Stichel, Reka Toth, Sarah Fritzsche, Moritz Anton Loeffler, Anna Melissa Schlitter, Olaf Neumann, Yassen Assenov, Monika Nadja Vogel, Arianeb Mehrabi, Katrin Hoffmann, Bruno Köhler, Christoph Springfeld, Dieter Weichenhan, Christoph Plass, Irene Esposito, Peter Schirmacher, Andreas von Deimling, Stephanie Roessler

Abstract

Objective: A detailed understanding of the molecular alterations in different forms of cholangiocarcinogenesis is crucial for a better understanding of cholangiocarcinoma (CCA) and may pave the way to early diagnosis and better treatment options.

Design: We analysed a clinicopathologically well-characterised patient cohort (n=54) with high-grade intraductal papillary (IPNB) or tubulopapillary (ITPN) neoplastic precursor lesions of the biliary tract and correlated the results with an independent non-IPNB/ITPN associated CCA cohort (n=294). The triplet sample set of non-neoplastic biliary epithelium, precursor and invasive CCA was analysed by next generation sequencing, DNA copy number and genome-wide methylation profiling.

Results: Patients with invasive CCA arising from IPNB/ITPN had better prognosis than patients with CCA not associated with IPNB/ITPN. ITPN was localised mostly intrahepatic, whereas IPNB was mostly of extrahepatic origin. IPNB/ITPN were equally associated with small-duct and large-duct type intrahepatic CCA. IPNB exhibited mutational profiles of extrahepatic CCA, while ITPN had significantly fewer mutations. Most mutations were shared between precursor lesions and corresponding invasive CCA but ROBO2 mutations occurred exclusively in invasive CCA and CTNNB1 mutations were mainly present in precursor lesions. In addition, IPNB and ITPN differed in their DNA methylation profiles and analyses of latent methylation components suggested that IPNB and ITPN may have different cells-of-origin.

Conclusion: Integrative analysis revealed that IPNB and ITPN harbour distinct early genetic alterations, IPNB are enriched in mutations typical for extrahepatic CCA, whereas ITPN exhibited few genetic alterations and showed distinct epigenetic profiles. In conclusion, IPNB/ITPN may represent a distinctive, intermediate form of intrahepatic and extrahepatic cholangiocarcinogenesis.

Keywords: bilary duct carcinoma; cholangiocarcinoma; gene mutation.

Conflict of interest statement

Competing interests: PS: Grant, boards and presentations from Novartis, and boards from Incyte. CP: Advisory board honoraria from BioMedX. BG: Advisory board from Novartis.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Study design, sample and patient characterisation. (A) A total of 54 patients with IPNB or ITPN neoplasms of the biliary tree were included in this study. The heatmap indicates the available DNA sequencing and DNA methylation/CNA data for IPNB/ITPN precursor lesions, invasive CCA and normal tissue. For 45 precursors lesions all three analyses, DNA sequencing, DNA methylation and CNA data were obtained. (B) For DNA sequencing, precursor lesions of 48 patients and 26 corresponding invasive CCA samples were analysed. (C) Genome-wide DNA methylation and genome-wide CNA profiling was performed for precursor lesions of 51 patients and compared with 41 corresponding non-neoplastic bile duct (normal) and 27 corresponding invasive CCA samples. (D) Immunohistochemical and histomorphological subtyping was performed for the entire cohort; immunohistochemical characterisation of one representative IPNB (top row) and one representative ITPN (bottom row) case is depicted. (E) Kaplan-Meier survival curves of IPNB and ITPN cases. (F) Kaplan-Meier survival curves of IPNB/ITPN-CCA cases with UICC2 or UICC3 compared with non-IPNB/ITPN-CCA cases of an independent cohort with matched numbers of UICC2 or UICC3, respectively. CCA, cholangiocarcinoma; CNA, copy number alteration; IPNB, intraductal papillary neoplasm of the bile duct; ITPN, intraductal tubulopapillary neoplasm of the bile duct; NA, not available.
Figure 2
Figure 2
Massive parallel sequencing reveals frequent genetic alterations in intraductal neoplasms of the bile duct. (A) Circos plot representing co-occurrence of mutations in all 48 non-invasive IPNB and ITPN samples. A band connecting genes represents co-occurring mutations in a given patient. The width of the band represents the frequency of this mutation pair within the dataset. (B) Oncoplot of genomic alterations in IPNB (n=39) and ITPN (n=9) samples. Missense mutations, in-frame mutations, truncations, amplificationsand deletions of 48 patients with intraductal neoplasms are shown. AMP, amplification; DEL, deletion; INFRAME, in-frame mutation; IPNB, intraductal papillary neoplasm of the bile duct; ITPN, intraductal tubulopapillary neoplasm of the bile duct; TRUNC, truncation.
Figure 3
Figure 3
IPNB/ITPN and corresponding invasive CCA tissue samples share genetic molecular profiles. (A) Oncoplot of genomic alterations in IPNB/ITPN (n=26) and (B) in the corresponding invasive CCA samples (n=26). Missense mutations, in-frame mutations, truncations, amplifications and deletions are shown. Genes (rows) of both oncoplots (A, B) were sorted by the total number of mutations in all samples. In addition, the order of samples (columns) in both oncoplots is based on the number of alterations in the precursor samples; thus, invasive CCA are sorted identical to the corresponding precursor lesions. (C) Overlap of specific mutations, that is, mutation of a certain amino acid in the respective gene, present in IPNB/ITPN precursor lesions and corresponding invasive CCA. (D) Shown are the proportions of mutations observed exclusively in the precursor lesions, exclusively in the invasive CCA or shared by the precursor lesion and corresponding invasive CCA in the indicated genes. (E) Diagram depicting the mutational alterations during progression from non-neoplastic normal bile to precursor and invasive CCA. AMP, amplification; DEL, deletion; INFRAME, in-frame mutation; IPNB, intraductal papillary neoplasm of the bile duct; ITPN, intraductal tubulopapillary neoplasm of the bile duct; ITPN, intraductal tubulopapillary neoplasm of the bile duct; TRUNC, truncation.
Figure 4
Figure 4
Copy number alteration profiles of normal, IPNB or ITPN and invasive CCA samples. (A) Copy number alterations of normal bile duct controls, (B) IPNB, (C) ITPN, (D) iCCA, (E) pCCA and (F) dCCA samples are shown along the chromosomes on the x-axes. The relative frequency of observed gains (green) and losses (red) are depicted above and below the horizontal line, respectively. CCA, cholangiocarcinoma; dCCA, distal CCA; iCCA, intrahepatic CCA; IPNB, intraductal papillary neoplasm of the bile duct; ITPN, intraductal tubulopapillary neoplasm of the bile duct; pCCA, perihilar CCA.
Figure 5
Figure 5
DNA methylation profiles of IPNB and ITPN precursor lesions. (A) Unsupervised hierarchical clustering analysis of the triplet normal bile duct, precursor and corresponding invasive CCA of 24 patients. The total number of samples is 72. (B) t-SNE plot of the DNA methylation profiles of bile duct controls of the corresponding IPNB, ITPN and ITPN-P cases (n=50), IPNB (n=41), ITPN (n=10) and ITPN-P (n=9). (C) MeDeCom analysis of IPNB (n=41), ITPN (n=10) and ITPN-P (n=9) samples and (D) of 27 IPNB and ITPN samples with corresponding invasive CCA. CCA, cholangiocarcinoma; IPNB, intraductal papillary neoplasm of the bile duct; ITPN, intraductal tubulopapillary neoplasm of the bile duct; ITPN-P, ITPN of the pancreas.

References

    1. Banales JM, Cardinale V, Carpino G, et al. . Expert consensus document: cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European network for the study of cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol 2016;13:261–80. 10.1038/nrgastro.2016.51
    1. Valle JW, Borbath I, Khan SA, et al. . Biliary cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2016;27:v28–37. 10.1093/annonc/mdw324
    1. Rizvi S, Khan SA, Hallemeier CL, et al. . Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol 2018;15:95–111. 10.1038/nrclinonc.2017.157
    1. Nagtegaal ID, Odze RD, Klimstra D, et al. . The 2019 who classification of tumours of the digestive system. Histopathology 2020;76:182–8. 10.1111/his.13975
    1. WHO Classification of Tumours Editorial Board . Who classification of tumours: digestive system tumours. 5th Edn. World Health Organization, 2019.
    1. Wan X-S, Xu Y-Y, Qian J-Y, et al. . Intraductal papillary neoplasm of the bile duct. World J Gastroenterol 2013;19:8595–604. 10.3748/wjg.v19.i46.8595
    1. Rocha FG, Lee H, Katabi N, et al. . Intraductal papillary neoplasm of the bile duct: a biliary equivalent to intraductal papillary mucinous neoplasm of the pancreas? Hepatology 2012;56:1352–60. 10.1002/hep.25786
    1. Yeh T-S, Tseng J-H, Chen T-C, et al. . Characterization of intrahepatic cholangiocarcinoma of the intraductal growth-type and its precursor lesions. Hepatology 2005;42:657–64. 10.1002/hep.20837
    1. Zen Y, Fujii T, Itatsu K, et al. . Biliary papillary tumors share pathological features with intraductal papillary mucinous neoplasm of the pancreas. Hepatology 2006;44:1333–43. 10.1002/hep.21387
    1. Zen Y, Sasaki M, Fujii T, et al. . Different expression patterns of mucin core proteins and cytokeratins during intrahepatic cholangiocarcinogenesis from biliary intraepithelial neoplasia and intraductal papillary neoplasm of the bile duct--an immunohistochemical study of 110 cases of hepatolithiasis. J Hepatol 2006;44:350–8. 10.1016/j.jhep.2005.09.025
    1. Klöppel G, Kosmahl M. Is the intraductal papillary mucinous neoplasia of the biliary tract a counterpart of pancreatic papillary mucinous neoplasm? J Hepatol 2006;44:249–50. 10.1016/j.jhep.2005.11.035
    1. Nakanuma Y, Kakuda Y, Uesaka K. Characterization of intraductal papillary neoplasm of the bile duct with respect to the histopathologic similarities to pancreatic intraductal papillary mucinous neoplasm. Gut Liver 2019;13:617–27. 10.5009/gnl18476
    1. Yang C-Y, Huang W-J, Tsai J-H, et al. . Targeted next-generation sequencing identifies distinct clinicopathologic and molecular entities of intraductal papillary neoplasms of the bile duct. Mod Pathol 2019;32:1637–45. 10.1038/s41379-019-0306-9
    1. Schlitter AM, Born D, Bettstetter M, et al. . Intraductal papillary neoplasms of the bile duct: stepwise progression to carcinoma involves common molecular pathways. Mod Pathol 2014;27:73–86. 10.1038/modpathol.2013.112
    1. Singhi AD, Wood LD, Parks E, et al. . Recurrent Rearrangements in PRKACA and PRKACB in Intraductal Oncocytic Papillary Neoplasms of the Pancreas and Bile Duct. Gastroenterology 2020;158:573–82. 10.1053/j.gastro.2019.10.028
    1. Moran S, Martínez-Cardús A, Sayols S, et al. . Epigenetic profiling to classify cancer of unknown primary: a multicentre, retrospective analysis. Lancet Oncol 2016;17:1386–95. 10.1016/S1470-2045(16)30297-2
    1. Capper D, Jones DTW, Sill M, et al. . Dna methylation-based classification of central nervous system tumours. Nature 2018;555:469–74. 10.1038/nature26000
    1. Koelsche C, Hartmann W, Schrimpf D, et al. . Array-Based DNA-methylation profiling in sarcomas with small blue round cell histology provides valuable diagnostic information. Mod Pathol 2018;31:1246–56. 10.1038/s41379-018-0045-3
    1. Torbenson M, Zen Y, Yeh MM. AFIP atlas of tumor pathology. Tumors of the Liver: ARP Press, 2018.
    1. Zen Y, Amarapurkar AD, Portmann BC. Intraductal tubulopapillary neoplasm of the bile duct: potential origin from peribiliary cysts. Hum Pathol 2012;43:440–5. 10.1016/j.humpath.2011.03.013
    1. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 2016;32:2847–9. 10.1093/bioinformatics/btw313
    1. Nakanuma Y, Jang K-T, Fukushima N, et al. . A statement by the Japan-Korea expert pathologists for future clinicopathological and molecular analyses toward consensus building of intraductal papillary neoplasm of the bile duct through several opinions at the present stage. J Hepatobiliary Pancreat Sci 2018;25:181–7. 10.1002/jhbp.532
    1. Nakanuma Y, Kakuda Y, Fukumura Y, et al. . The pathologic and genetic characteristics of the intestinal subtype of intraductal papillary neoplasms of the bile duct. Am J Surg Pathol 2019;43:1212–20. 10.1097/PAS.0000000000001295
    1. Aoki Y, Mizuma M, Hata T, et al. . Intraductal papillary neoplasms of the bile duct consist of two distinct types specifically associated with clinicopathological features and molecular phenotypes. J Pathol 2020;251:38–48. 10.1002/path.5398
    1. Schlitter AM, Jang K-T, Klöppel G, et al. . Intraductal tubulopapillary neoplasms of the bile ducts: clinicopathologic, immunohistochemical, and molecular analysis of 20 cases. Mod Pathol 2016;29:93. 10.1038/modpathol.2015.106
    1. Wardell CP, Fujita M, Yamada T, et al. . Genomic characterization of biliary tract cancers identifies driver genes and predisposing mutations. J Hepatol 2018;68:959–69. 10.1016/j.jhep.2018.01.009
    1. Chaisaingmongkol J, Budhu A, Dang H, et al. . Common molecular subtypes among Asian hepatocellular carcinoma and cholangiocarcinoma. Cancer Cell 2017;32:57–70. 10.1016/j.ccell.2017.05.009
    1. Farshidfar F, Zheng S, Gingras M-C, et al. . Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-Mutant molecular profiles. Cell Rep 2017;18:2780–94. 10.1016/j.celrep.2017.02.033
    1. Jusakul A, Cutcutache I, Yong CH, et al. . Whole-Genome and epigenomic landscapes of etiologically distinct subtypes of cholangiocarcinoma. Cancer Discov 2017;7:1116–35. 10.1158/-17-0368
    1. Goeppert B, Toth R, Singer S, et al. . Integrative analysis defines distinct prognostic subgroups of intrahepatic cholangiocarcinoma. Hepatology 2019;69:2091–106. 10.1002/hep.30493
    1. Lutsik P, Slawski M, Gasparoni G, et al. . MeDeCom: discovery and quantification of latent components of heterogeneous methylomes. Genome Biol 2017;18:55. 10.1186/s13059-017-1182-6
    1. Luvira V, Pugkhem A, Bhudhisawasdi V, et al. . Long-Term outcome of surgical resection for intraductal papillary neoplasm of the bile duct. J Gastroenterol Hepatol 2017;32:527–33. 10.1111/jgh.13481
    1. Nakamura H, Arai Y, Totoki Y, et al. . Genomic spectra of biliary tract cancer. Nat Genet 2015;47:1003–10. 10.1038/ng.3375
    1. Goeppert B, Folseraas T, Roessler S, et al. . Genomic characterization of cholangiocarcinoma in primary sclerosing cholangitis reveals therapeutic opportunities. Hepatology 2020;72:1253–66. 10.1002/hep.31110
    1. Albrecht T, Rausch M, Rössler S, et al. . Her2 gene (ErbB2) amplification is a rare event in non-liver-fluke associated cholangiocarcinogenesis. BMC Cancer 2019;19:1191. 10.1186/s12885-019-6320-y
    1. Goeppert B, Ernst C, Baer C, et al. . Cadherin-6 is a putative tumor suppressor and target of epigenetically dysregulated miR-429 in cholangiocarcinoma. Epigenetics 2016;11:780–90. 10.1080/15592294.2016.1227899
    1. Wang J, Wang H, Peters M, et al. . Loss of FBXW7 synergizes with activated Akt signaling to promote c-myc dependent cholangiocarcinogenesis. J Hepatol 2019;71:742–52. 10.1016/j.jhep.2019.05.027
    1. Dickson BJ, Gilestro GF. Regulation of commissural axon pathfinding by slit and its Robo receptors. Annu Rev Cell Dev Biol 2006;22:651–75. 10.1146/annurev.cellbio.21.090704.151234
    1. Ong CK, Subimerb C, Pairojkul C, et al. . Exome sequencing of liver fluke-associated cholangiocarcinoma. Nat Genet 2012;44:690–3. 10.1038/ng.2273
    1. Kim Y-H, Hong E-K, Kong S-Y, et al. . Two classes of intrahepatic cholangiocarcinoma defined by relative abundance of mutations and copy number alterations. Oncotarget 2016;7:23825–36. 10.18632/oncotarget.8183
    1. Zhao S-J, Shen Y-F, Li Q, et al. . Slit2/Robo1 axis contributes to the Warburg effect in osteosarcoma through activation of SRC/ERK/c-MYC/PFKFB2 pathway. Cell Death Dis 2018;9:390. 10.1038/s41419-018-0419-y
    1. Dallol A, Morton D, Maher ER, et al. . Slit2 axon guidance molecule is frequently inactivated in colorectal cancer and suppresses growth of colorectal carcinoma cells. Cancer Res 2003;63:1054–8.
    1. Huang Z, Wen P, Kong R, et al. . Usp33 mediates slit-robo signaling in inhibiting colorectal cancer cell migration. Int J Cancer 2015;136:1792–802. 10.1002/ijc.29226
    1. Tseng R-C, Lee S-H, Hsu H-S, et al. . Slit2 attenuation during lung cancer progression deregulates beta-catenin and E-cadherin and associates with poor prognosis. Cancer Res 2010;70:543–51. 10.1158/0008-5472.CAN-09-2084
    1. Vaughen J, Igaki T. Slit-Robo repulsive signaling Extrudes tumorigenic cells from epithelia. Dev Cell 2016;39:683–95. 10.1016/j.devcel.2016.11.015
    1. Fujikura K, Akita M, Ajiki T, et al. . Recurrent mutations in APC and CTNNB1 and activated Wnt/β-catenin signaling in intraductal papillary neoplasms of the bile duct: a whole exome sequencing study. Am J Surg Pathol 2018;42:1674–85. 10.1097/PAS.0000000000001155
    1. Goeppert B, Konermann C, Schmidt CR, et al. . Global alterations of DNA methylation in cholangiocarcinoma target the Wnt signaling pathway. Hepatology 2014;59:544–54. 10.1002/hep.26721
    1. Sugimachi K, Taguchi K, Aishima S, et al. . Altered expression of beta-catenin without genetic mutation in intrahepatic cholangiocarcinoma. Mod Pathol 2001;14:900–5. 10.1038/modpathol.3880409
    1. Sasaki M, Matsubara T, Nitta T, et al. . Gnas and KRAS mutations are common in intraductal papillary neoplasms of the bile duct. PLoS One 2013;8:e81706. 10.1371/journal.pone.0081706

Source: PubMed

3
Subskrybuj