Base excision repair deficiency signatures implicate germline and somatic MUTYH aberrations in pancreatic ductal adenocarcinoma and breast cancer oncogenesis

My Linh Thibodeau, Eric Y Zhao, Caralyn Reisle, Carolyn Ch'ng, Hui-Li Wong, Yaoqing Shen, Martin R Jones, Howard J Lim, Sean Young, Carol Cremin, Erin Pleasance, Wei Zhang, Robert Holt, Peter Eirew, Joanna Karasinska, Steve E Kalloger, Greg Taylor, Elisa Majounie, Melika Bonakdar, Zusheng Zong, Dustin Bleile, Readman Chiu, Inanc Birol, Karen Gelmon, Caroline Lohrisch, Karen L Mungall, Andrew J Mungall, Richard Moore, Yussanne P Ma, Alexandra Fok, Stephen Yip, Aly Karsan, David Huntsman, David F Schaeffer, Janessa Laskin, Marco A Marra, Daniel J Renouf, Steven J M Jones, Kasmintan A Schrader, My Linh Thibodeau, Eric Y Zhao, Caralyn Reisle, Carolyn Ch'ng, Hui-Li Wong, Yaoqing Shen, Martin R Jones, Howard J Lim, Sean Young, Carol Cremin, Erin Pleasance, Wei Zhang, Robert Holt, Peter Eirew, Joanna Karasinska, Steve E Kalloger, Greg Taylor, Elisa Majounie, Melika Bonakdar, Zusheng Zong, Dustin Bleile, Readman Chiu, Inanc Birol, Karen Gelmon, Caroline Lohrisch, Karen L Mungall, Andrew J Mungall, Richard Moore, Yussanne P Ma, Alexandra Fok, Stephen Yip, Aly Karsan, David Huntsman, David F Schaeffer, Janessa Laskin, Marco A Marra, Daniel J Renouf, Steven J M Jones, Kasmintan A Schrader

Abstract

We report a case of early-onset pancreatic ductal adenocarcinoma in a patient harboring biallelic MUTYH germline mutations, whose tumor featured somatic mutational signatures consistent with defective MUTYH-mediated base excision repair and the associated driver KRAS transversion mutation p.Gly12Cys. Analysis of an additional 730 advanced cancer cases (N = 731) was undertaken to determine whether the mutational signatures were also present in tumors from germline MUTYH heterozygote carriers or if instead the signatures were only seen in those with biallelic loss of function. We identified two patients with breast cancer each carrying a pathogenic germline MUTYH variant with a somatic MUTYH copy loss leading to the germline variant being homozygous in the tumor and demonstrating the same somatic signatures. Our results suggest that monoallelic inactivation of MUTYH is not sufficient for C:G>A:T transversion signatures previously linked to MUTYH deficiency to arise (N = 9), but that biallelic complete loss of MUTYH function can cause such signatures to arise even in tumors not classically seen in MUTYH-associated polyposis (N = 3). Although defective MUTYH is not the only determinant of these signatures, MUTYH germline variants may be present in a subset of patients with tumors demonstrating elevated somatic signatures possibly suggestive of MUTYH deficiency (e.g., COSMIC Signature 18, SigProfiler SBS18/SBS36, SignatureAnalyzer SBS18/SBS36).

Trial registration: ClinicalTrials.gov NCT02155621.

Keywords: neoplasm of the breast; neoplasm of the pancreas; pancreatic adenocarcinoma.

© 2019 Thibodeau et al.; Published by Cold Spring Harbor Laboratory Press.

Figures

Figure 1.
Figure 1.
Patient #1 compound heterozygous MUTYH germline variants. (A) Integrated Genomics Viewer (IGV) (Thorvaldsdóttir et al. 2013) capture of whole-genome sequencing of tumor and matched blood DNA with whole-transcriptome sequencing of tumor RNA data at the genomic region encompassing the two MUTYH variants (c.996G>A, p.Ser332Ser and c.815G>A, p.Gly272Glu; NM_001048171). Paired-end transcriptome reads showing the germline variants to be in trans with read pairs containing the p.Gly272Glu variant (exon 10) but not the p.Ser332Ser variant (exon 12) (red box). The splicing aberration caused by p.Ser332Ser variant was not visually apparent in IGV and required transcriptome TAP for detection and characterization. TAP was used to assemble reads into contigs. Contig 1 shows aberrant splicing and contig 2 shows normal splicing. (B) Schematic of TAP analysis results showing the synonymous germline variant (c.996G>A, p.Ser332Ser) creating a novel canonical AG acceptor splice site at Chr 1:45797480–45797481 and removing 42 bp (14 amino acids).
Figure 2.
Figure 2.
Comparison of mutation signatures exposures of MUTYH germline or combined germline/somatic biallelic aberrations against 731 cancer genomes of mixed origins. The cohort distribution of mutation signature exposures for each signature is shown using signature composition reference matrices from COSMIC, SigProfiler, and SignatureAnalyzer. The mutation fraction exposures (proportion of all mutations contributed by each signature) of Patient #1, Patient #2, and Patient #3 are superimposed. Signatures previously associated with MUTYH-mediated BER deficiency are highlighted by a rectangle. Patient #1 (PDAC, pink diamond), Patient #2 (breast cancer, green diamond), and Patient #3 (breast cancer, blue diamond) demonstrated germline or combined germline/somatic biallelic MUTYH aberrations. Functional biallelic MUTYH loss of function was present in tumors of Patient #1, Patient #2, and Patient #3 and all three patients displayed elevated outlier (>1.5 interquartile range above third quartile) signatures previously associated with defective MUTYH (COSMIC Signature 18, SigProfiler SBS18 and SBS36, and SignatureAnalyzer SBS18 and SBS36).
Figure 3.
Figure 3.
Mutation catalogs and COSMIC mutational signatures of MUTYH germline or combined germline/somatic biallelic aberrations. Complete catalogs of SNVs from WGS of tumors were classified based on variant and 3′/5′ contexts into 96 categories (left side) and mutation catalogs were used to calculate the contribution of each signature to mutational burden (right side). The proportion of mutations or mutation count in each of the 96 categories is shown here as a barplot (left side), whereas the signIT Markov Chain Monte Carlo simulation result for COSMIC mutational signatures is shown as a violin plot distribution (right side). COSMIC Signature 18 composition is shown at the top for reference. Patient #1 (germline MUTYH c.815G>A, p.Gly272Glu; c.996G>A, p.Ser332Ser; NM_001048171) and Patient #2 (germline MUTYH c.892-2A>G) tumors showed increased Signature 18 somatic mutation burden, as exemplified by the resemblance of their mutation catalogs to COSMIC reference Signature 18. Other mutational processes contributed most to the mutational burden of Patient #3's tumor.
Figure 4.
Figure 4.
Germline MUTYH founder Asian splice site variant impact on splicing. The Asian founder mutation MUTYH disrupting the canonical acceptor splice site at exon 11 (c.892-2A>G) was found in six patients (Patient #2, Patient #6, Patient #9, Patient #10, Patient #11, and Patient #12). (A) Integrated Genomic Viewer (IGV) (Thorvaldsdóttir et al. 2013) capture of whole-genome sequencing of tumor and matched blood DNA with whole-transcriptome sequencing of tumor RNA data at the genomic region encompassing the MUTYH variant (c.892-2A>G; NM_001048171).Transcriptome data showing abnormal splicing removing 9 bp from exon 11 (19 reads with a black line) and marked intron 10 and intron 11 retention. (B) Schematic of abnormal splicing removing 9 bp from exon 11. (C) Schematic of abnormal splicing resulting in intron retention.

References

    1. Aimé A, Coulet F, Lefevre JH, Colas C, Cervera P, Flejou J-F, Lascols O, Soubrier F, Parc Y. 2015. Somatic c.34G>T KRAS mutation: a new prescreening test for MUTYH-associated polyposis? Cancer Genet 208: 390–395. 10.1016/j.cancergen.2015.04.005
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SAJR, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Børresen-Dale A-L, et al. 2013. Signatures of mutational processes in human cancer. Nature 500: 415–421. 10.1038/nature12477
    1. Alexandrov L, Kim J, Haradhvala NJ, Huang MN, Ng AW, Boot A, Covington KR, Gordenin DA, Bergstrom E, Lopez-Bigas N, et al. 2018. The repertoire of mutational signatures in human cancer. bioRxiv 1: 1–27. 10.1101/322859
    1. Aretz S, Tricarico R, Papi L, Spier I, Pin E, Horpaopan S, Cordisco EL, Pedroni M, Stienen D, Gentile A, et al. 2013. MUTYH-associated polyposis (MAP): evidence for the origin of the common European mutations p.Tyr179Cys and p.Gly396Asp by founder events. Eur J Hum Genet 22: 923–929. 10.1038/ejhg.2012.309.
    1. Bailey P, Chang DK, Nones K, Johns AL, Patch A-M, Gingras M-C, Miller DK, Christ AN, Bruxner TJC, Quinn MC, et al. 2016. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 531: 47–52. 10.1038/nature16965
    1. Boesaard EP, Vogelaar IP, Bult P, Wauters CA, van Krieken JHJ, Ligtenberg MJ, van der Post RS, Hoogerbrugge N. 2014. Germline MUTYH gene mutations are not frequently found in unselected patients with papillary breast carcinoma. Hered Cancer Clin Pract 12: 21 10.1186/1897-4287-12-21
    1. Cheadle JP, Sampson JR. 2007. MUTYH-associated polyposis—from defect in base excision repair to clinical genetic testing. DNA Repair 6: 274–279. 10.1016/j.dnarep.2006.11.001
    1. Cheng KC, Cahill DS, Kasai H, Nishimura S, Loeb LA. 1992. 8-Hydroxyguanine, an abundant form of oxidative DNA damage, causes G->T and A->C substitutions. J Biol Chem 267: 166–172.
    1. Chiu R, Nip KM, Chu J, Birol I. 2018. TAP: a targeted clinical genomics pipeline for detecting transcript variants using RNA-seq data. BMC Med Genomics 11: 7–9. 10.1186/s12920-018-0402-6
    1. Egashira A, Yamauchi K, Yoshiyama K, Kawate H, Katsuki M, Sekiguchi M, Sugimachi K, Maki H, Tsuzuki T. 2002. Mutational specificity of mice defective in the MTH1 and/or the MSH2 genes. DNA Repair 1: 881–893. 10.1016/S1568-7864(02)00113-1
    1. Fokkema IFAC, Taschner PEM, Schaafsma GCP, Celli J, Laros JFJ, den Dunnen JT. 2011. LOVD v.2.0: the next generation in gene variant databases. Hum Mutat 32: 557–563. 10.1002/humu.21438
    1. Forbes SA, Beare D, Boutselakis H, Bamford S, Bindal N, Tate J, Cole CG, Ward S, Dawson E, Ponting L, et al. 2017. COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 45: D777–D783. 10.1093/nar/gkw1121
    1. Ha G, Roth A, Lai D, Bashashati A, Ding J, Goya R, Giuliany R, Rosner J, Oloumi A, Shumansky K, et al. 2012. Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer. Genome Res 22: 1995–2007. 10.1101/gr.137570.112
    1. Haradhvala NJ, Kim J, Maruvka YE, Polak P, Rosebrock D, Livitz D, Hess JM, Leshchiner I, Kamburov A, Mouw KW, et al. 2018. Distinct mutational signatures characterize concurrent loss of polymerase proofreading and mismatch repair. Nat Commun 9: 1–9. 10.1038/s41467-018-04002-4
    1. Jones S, Lambert S, Williams GT, Best JM, Sampson JR, Cheadle JP. 2004. Increased frequency of the k-ras G12C mutation in MYH polyposis colorectal adenomas. Br J Cancer 90: 1591–1593. 10.1038/sj.bjc.6601747
    1. Jones SJ, Laskin J, Li YY, Griffith OL, An J, Bilenky M, Butterfield YS, Cezard T, Chuah E, Corbett R, et al. 2010. Evolution of an adenocarcinoma in response to selection by targeted kinase inhibitors. Genome Biol 11: R82 10.1186/gb-2010-11-8-r82
    1. Kairupan CF, Meldrum CJ, Crooks R, Milward EA, Spigelman AD, Burgess B, Groombridge C, Kirk J, Tucker K, Ward R, et al. 2005. Mutation analysis of the MYH gene in an Australian series of colorectal polyposis patients with or without germline APC mutations. Int J Cancer 116: 73–77. 10.1002/ijc.20983
    1. Klungland A, Rosewell I, Hollenbach S, Larsen E, Daly G, Epe B, Seeberg E, Lindahl T, Barnes DE. 1999. Accumulation of premutagenic DNA lesions in mice defective in removal of oxidative base damage. Proc Natl Acad Sci 96: 13300–13305. 10.1073/pnas.96.23.13300
    1. Landrum MJ, Lee JM, Benson M, Brown G, Chao C, Chitipiralla S, Gu B, Hart J, Hoffman D, Hoover J, et al. 2016. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res 44: D862–D868. 10.1093/nar/gkv1222
    1. Lejbkowicz F, Cohen I, Barnett-Griness O, Pinchev M, Poynter J, Gruber SB, Rennert G. 2012. Common MUTYH mutations and colorectal cancer risk in multiethnic populations. Fam Cancer 11: 329–335. 10.1007/s10689-012-9516-8
    1. Lek M, Karczewski KJ, Minikel EV, Samocha KE, Banks E, Fennell T, O'Donnell-Luria AH, Ware JS, Hill AJ, Cummings BB, et al. 2016. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536: 285–291. 10.1038/nature19057
    1. Minowa O, Arai T, Hirano M, Monden Y, Nakai S, Fukuda M, Itoh M, Takano H, Hippou Y, Aburatani H, et al. 2000. Mmh/Ogg1 gene inactivation results in accumulation of 8-hydroxyguanine in mice. Proc Natl Acad Sci 97: 4156–4161. 10.1073/pnas.050404497
    1. Mur P, Jemth A-S, Bevc L, Amaral N, Navarro M, Valdés-Mas R, Pons T, Aiza G, Urioste M, Valencia A, et al. 2018. Germline variation in the oxidative DNA repair genes NUDT1 and OGG1 is not associated with hereditary colorectal cancer or polyposis. Hum Mutat 39: 1214–1225. 10.1002/humu.23564
    1. Pilati C, Shinde J, Alexandrov LB, Assié G, André T, Hélias-Rodzewicz Z, Ducoudray R, Le Corre D, Zucman-Rossi J, Emile J-F, et al. 2017. Mutational signature analysis identifies MUTYH deficiency in colorectal cancers and adrenocortical carcinomas. J Pathol 242: 10–15. 10.1002/path.4880
    1. Romanel A, Zhang T, Elemento O, Demichelis F. 2017. EthSEQ: ethnicity annotation from whole exome sequencing data. Bioinformatics 33: 2402–2404. 10.1093/bioinformatics/btx165
    1. Sakai A, Nakanishi M, Yoshiyama K, Maki H. 2006. Impact of reactive oxygen species on spontaneous mutagenesis in Escherichia coli. Genes Cells 11: 767–778. 10.1111/j.1365-2443.2006.00982.x
    1. Sakamoto K, Tominaga Y, Yamauchi K, Nakatsu Y, Sakumi K, Yoshiyama K, Egashira A, Kura S, Yao T, Tsuneyoshi M, et al. 2007. MUTYH-null mice are susceptible to spontaneous and oxidative stress–induced intestinal tumorigenesis. Cancer Res 67: 6599–6604. 10.1158/0008-5472.CAN-06-4802
    1. Sakumi K, Tominaga Y, Furuichi M, Xu P, Tsuzuki T, Sekiguchi M, Nakabeppu Y. 2003. Ogg1 knockout-associated lung tumorigenesis and its suppression by Mth1 gene disruption. Cancer Res 63: 902–905.
    1. Saunders CT, Wong WSW, Swamy S, Becq J, Murray LJ, Cheetham RK. 2012. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 28: 1811–1817. 10.1093/bioinformatics/bts271
    1. Scarpa A, Chang DK, Nones K, Corbo V, Patch A-M, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, et al. 2017. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543: 65–71. 10.1038/nature21063
    1. Smith LM, Sharif S, Brand R, Fink E, Lamb J, Whitcomb DC. 2009. MUTYH exon 7 and 13 mutations associated with colorectal cancer (MAP syndrome) are not commonly associated with sporadic pancreatic cancer. Pancreatology 9: 793–796. 10.1159/000199440
    1. Suzuki T, Harashima H, Kamiya H. 2010. Effects of base excision repair proteins on mutagenesis by 8-oxo-7,8-dihydroguanine (8-hydroxyguanine) paired with cytosine and adenine. DNA Repair 9: 542–550. 10.1016/j.dnarep.2010.02.004
    1. Taki K, Sato Y, Nomura S, Ashihara Y, Kita M, Tajima I, Sugano K, Arai M. 2015. Mutation analysis of MUTYH in Japanese colorectal adenomatous polyposis patients. Fam Cancer 15: 261–265. 10.1007/s10689-015-9857-1
    1. Thibodeau ML, Zhao E, Reisle C, Zhang W, Thiessen N, Bleile D, Mungall AJ, Ma YP, Renouf DJ, Ng T, et al. 2018. Whole genome and whole transcriptome genomic profiling of a metastatic eccrine porocarcinoma. NPJ Precis Oncol 2: 8 10.1038/s41698-018-0050-5
    1. Thorvaldsdóttir H, Robinson JT, Mesirov JP. 2013. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14: 178–192. 10.1093/bib/bbs017
    1. Tsuzuki T, Egashira A, Igarashi H, Iwakuma T, Nakatsuru Y, Tominaga Y, Kawate H, Nakao K, Nakamura K, Ide F, et al. 2001. Spontaneous tumorigenesis in mice defective in the MTH1 gene encoding 8-oxo-dGTPase. Proc Natl Acad Sci 98: 11456–11461. 10.1073/pnas.191086798
    1. Viel A, Bruselles A, Meccia E, Fornasarig M, Quaia M, Canzonieri V, Policicchio E, Urso ED, Agostini M, Genuardi M, et al. 2017. A specific mutational signature associated with DNA 8-oxoguanine persistence in MUTYH-defective colorectal cancer. EBioMedicine 20: 39–49. 10.1016/j.ebiom.2017.04.022
    1. Wasielewski M, Out AA, Vermeulen J, Nielsen M, van den Ouweland A, Tops CMJ, Wijnen JT, Vasen HFA, Weiss MM, Klijn JGM, et al. 2010. Increased MUTYH mutation frequency among Dutch families with breast cancer and colorectal cancer. Breast Cancer Res Treat 124: 635–641. 10.1007/s10549-010-0801-7
    1. Weren RDA, Ligtenberg MJL, Kets CM, de Voer RM, Verwiel ETP, Spruijt L, van Zelst-Stams WAG, Jongmans MC, Gilissen C, Hehir Kwa JY, et al. 2015. A germline homozygous mutation in the base-excision repair gene NTHL1 causes adenomatous polyposis and colorectal cancer. Nat Genet 47: 668–671. 10.1038/ng.3287
    1. Win AK, Cleary SP, Dowty JG, Baron JA, Young JP, Buchanan DD, Southey MC, Burnett T, Parfrey PS, Green RC, et al. 2011. Cancer risks for monoallelic MUTYH mutation carriers with a family history of colorectal cancer. Int J Cancer 129: 2256–2262. 10.1002/ijc.25870
    1. Zhao EY, Shen Y, Pleasance E, Kasaian K, Leelakumari S, Jones M, Bose P, Ch'ng C, Reisle C, Eirew P, et al. 2017. Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer. Clin Cancer Res 23: 7521–7530. 10.1158/1078-0432.CCR-17-1941
    1. Zhou L, Baba Y, Kitano Y, Miyake K, Zhang X, Yamamura K, Kosumi K, Kaida T, Arima K, Taki K, et al. 2016. KRAS, BRAF, and PIK3CA mutations, and patient prognosis in 126 pancreatic cancers: pyrosequencing technology and literature review. Med Oncol 33: 32 10.1007/s12032-016-0745-9

Source: PubMed

3
Abonnieren