Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The context-specific role of germline pathogenicity in tumorigenesis

Abstract

Human cancers arise from environmental, heritable and somatic factors, but how these mechanisms interact in tumorigenesis is poorly understood. Studying 17,152 prospectively sequenced patients with cancer, we identified pathogenic germline variants in cancer predisposition genes, and assessed their zygosity and co-occurring somatic alterations in the concomitant tumors. Two major routes to tumorigenesis were apparent. In carriers of pathogenic germline variants in high-penetrance genes (5.1% overall), lineage-dependent patterns of biallelic inactivation led to tumors exhibiting mechanism-specific somatic phenotypes and fewer additional somatic oncogenic drivers. Nevertheless, 27% of cancers in these patients, and most tumors in patients with pathogenic germline variants in lower-penetrance genes, lacked particular hallmarks of tumorigenesis associated with the germline allele. The dependence of tumors on pathogenic germline variants is variable and often dictated by both penetrance and lineage, a finding with implications for clinical management.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Germline pathogenicity in prospectively characterized advanced cancers.
Fig. 2: Penetrance and tumor lineage drive selection for somatic biallelic inactivation.
Fig. 3: Phenotypic and evolutionary consequences of biallelic inactivation high-penetrance alleles.
Fig. 4: Lineage and zygosity-dependent somatic phenotypes of germline MMR carriers.
Fig. 5: Dispensability of germline pathogenicity in tumorigenesis.
Fig. 6: Integration of germline and somatic tumor profiling.

Similar content being viewed by others

Data availability

Study results including cohort-wide prevalence and zygosity of germline and somatic mutations are available at https://www.signaldb.org and may be subject to a registration process and certain terms of use specified at https://www.signaldb.org/terms, including that the results may be used only for noncommercial research purposes without a license agreement with MSK Cancer Center. Germline variants and tumor-specific zygosity estimates are available from the National Center for Biotechnology Information dbGaP archive at accession no. phs001858.v1.p1. In addition, the following publicly available data were used: annotations indicating statistically significant somatic mutations were derived from Hotspots (http://www.cancerhotspots.org); biological effects, prognostic information and treatment implications of specific cancer gene alterations were obtained from OncoKB as of June 2018 (http://www.oncokb.org); variant-level annotations aggregated from data resources for germline alterations were obtained from myvariant.info as of August 2017 (https://myvariant.info); population frequencies for observed germline alterations were derived from gnomAD r2.0.2 (https://gnomad.broadinstitute.org); and annotations regarding the deleterious nature of known germline variants and associated phenotypes were downloaded from ClinVar as of September 2017 (https://www.ncbi.nlm.nih.gov/clinvar).

Code availability

Source code is available at https://github.com/taylor-lab/somatic-germline.

References

  1. Garber, J. E. & Offit, K. Hereditary cancer predisposition syndromes. J. Clin. Oncol. 23, 276–292 (2005).

    Article  PubMed  Google Scholar 

  2. Rahman, N. Realizing the promise of cancer predisposition genes. Nature 505, 302–308 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Knudson, A. G. Mutation and cancer: statistical study of retinoblastoma. Proc. Natl Acad. Sci. USA 68, 820–823 (1971).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Nichols, K. E., Malkin, D., Garber, J. E., Fraumeni, J. F. & Li, F. P. Germ-line p53 mutations predispose to a wide spectrum of early-onset cancers. Cancer Epidemiol. Biomark. Prev. 10, 83–87 (2001).

    CAS  Google Scholar 

  5. Chandrasekharappa, S. C. et al. Positional cloning of the gene for multiple endocrine neoplasia-type 1. Science 276, 404–407 (1997).

    Article  CAS  PubMed  Google Scholar 

  6. Moore, K. et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N. Engl. J. Med. 379, 2495–2505 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fong, P. C. et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 361, 123–134 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Stadler, Z. K., Schrader, K. A., Vijai, J., Robson, M. E. & Offit, K. Cancer genomics and inherited risk. J. Clin. Oncol. 32, 687–698 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tung, N. et al. Counselling framework for moderate-penetrance cancer-susceptibility mutations. Nat. Rev. Clin. Oncol. 13, 581–588 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jasperson, K. W., Tuohy, T. M., Neklason, D. W. & Burt, R. W. Hereditary and familial colon cancer. Gastroenterology 138, 2044–2058 (2010).

    Article  CAS  PubMed  Google Scholar 

  12. Hampel, H. & Peltomaki, P. Hereditary colorectal cancer: risk assessment and management. Clin. Genet. 58, 89–97 (2000).

    Article  CAS  PubMed  Google Scholar 

  13. Couch, F. J. et al. Associations between cancer predisposition testing panel genes and breast cancer. JAMA Oncol. 3, 1190–1196 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Thavaneswaran, S. et al. Therapeutic implications of germline genetic findings in cancer. Nat. Rev. Clin. Oncol. 16, 386–396 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Zhang, J. et al. Germline mutations in predisposition genes in pediatric cancer. N. Engl. J. Med. 373, 2336–2346 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Huang, K.-L. et al. Pathogenic germline variants in 10,389 adult cancers. Cell 173, 355–370.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lu, C. et al. Patterns and functional implications of rare germline variants across 12 cancer types. Nat. Commun. 6, 10086 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Mandelker, D. et al. Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA vs guideline-based germline testing. JAMA 318, 825–835 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Jonsson, P. et al. Tumour lineage shapes BRCA-mediated phenotypes. Nature 571, 576–579 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cheng, D. T. et al. Comprehensive detection of germline variants by MSK-IMPACT, a clinical diagnostic platform for solid tumor molecular oncology and concurrent cancer predisposition testing. BMC Med. Genom. 10, 33 (2017).

    Article  Google Scholar 

  21. Richards, S. et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet. Med. 17, 405–424 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Gryfe, R., Di Nicola, N., Gallinger, S. & Redston, M. Somatic instability of the APC I1307K allele in colorectal neoplasia. Cancer Res. 58, 4040–4043 (1998).

    CAS  PubMed  Google Scholar 

  23. Win, A. K., Hopper, J. L. & Jenkins, M. A. Association between monoallelic MUTYH mutation and colorectal cancer risk: a meta-regression analysis. Fam. Cancer 10, 1–9 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Knudson, A. G. Two genetic hits (more or less) to cancer. Nat. Rev. Cancer 1, 157–162 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Nickerson, M. L. et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt–Hogg–Dubé syndrome. Cancer Cell 2, 157–164 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Polak, P. et al. A mutational signature reveals alterations underlying deficient homologous recombination repair in breast cancer. Nat. Genet. 49, 1476–1486 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Pilati, C. et al. Mutational signature analysis identifies MUTYH deficiency in colorectal cancers and adrenocortical carcinomas. J. Pathol. 242, 10–15 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Viel, A. et al. A specific mutational signature associated with DNA 8-oxoguanine persistence in MUTYH-defective colorectal cancer. eBioMedicine 20, 39–49 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Scarpa, A. et al. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature 543, 65–71 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Chakravarty, D. et al. Oncokb: a precision oncology knowledge base. JCO Precis. Oncol. https://doi.org/10.1200/PO.17.00011 (2017).

  32. Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409–413 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lynch, H. T., Shaw, M. W., Magnuson, C. W., Larsen, A. L. & Krush, A. J. Hereditary factors in cancer. Study of two large midwestern kindreds. Arch. Intern. Med. 117, 206–212 (1966).

    Article  CAS  PubMed  Google Scholar 

  34. Lynch, H. T. & Krush, A. J. Cancer family ‘G’ revisited: 1895–1970. Cancer 27, 1505–1511 (1971).

    Article  CAS  PubMed  Google Scholar 

  35. Latham, A. et al. Microsatellite instability is associated with the presence of Lynch syndrome pan-cancer. J. Clin. Oncol. 37, 286–295 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Møller, P. et al. Cancer risk and survival in path_MMR carriers by gene and gender up to 75 years of age: a report from the Prospective Lynch Syndrome Database. Gut 67, 1306–1316 (2018).

    Article  PubMed  Google Scholar 

  37. Kinzler, K. W. & Vogelstein, B. Landscaping the cancer terrain. Science 280, 1036–1037 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Kinzler, K. W. & Vogelstein, B. Cancer-susceptibility genes. Gatekeepers and caretakers. Nature 386, 761–763 (1997).

    Article  CAS  PubMed  Google Scholar 

  39. Baysal, B. E. et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 287, 848–851 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Karaayvaz-Yildirim, M. et al. Aneuploidy and a deregulated DNA damage response suggest haploinsufficiency in breast tissues of BRCA2 mutation carriers. Sci. Adv. 6, eaay2611 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Cheng, D. T. et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J. Mol. Diagn. 17, 251–264 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Won, H. H., Scott, S. N., Brannon, A. R., Shah, R. H. & Berger, M. F. Detecting somatic genetic alterations in tumor specimens by exon capture and massively parallel sequencing. J. Vis. Exp. 80, e50710 (2013).

    Google Scholar 

  43. Xin, J. et al. High-performance web services for querying gene and variant annotation. Genome Biol. 17, 91 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Karczewski, K. J. et al. The mutational constraint spectrum quantified from variation in 141,456 humans. Nature 581, 434–443 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Landrum, M. J. et al. ClinVar: public archive of interpretations of clinically relevant variants. Nucleic Acids Res. 44, D862–D868 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Nakagawa, H. et al. Mismatch repair gene PMS2: disease-causing germline mutations are frequent in patients whose tumors stain negative for PMS2 protein, but paralogous genes obscure mutation detection and interpretation. Cancer Res. 64, 4721–4727 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Hayward, B. E. et al. Extensive gene conversion at the PMS2 DNA mismatch repair locus. Hum. Mutat. 28, 424–430 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Kilpivaara, O., Alhopuro, P., Vahteristo, P., Aaltonen, L. A. & Nevanlinna, H. CHEK2 I157T associates with familial and sporadic colorectal cancer. J. Med. Genet. 43, e34 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kilpivaara, O. et al. CHEK2 variant I157T may be associated with increased breast cancer risk. Int. J. Cancer 111, 543–547 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Apostolou, P. & Papasotiriou, I. Current perspectives on CHEK2 mutations in breast cancer. Breast Cancer 9, 331–335 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, L. et al. Fumarate hydratase FH c.1431_1433dupAAA (p.Lys477dup) variant is not associated with cancer including renal cell carcinoma. Hum. Mutat. 41, 103–109 (2020).

    Article  PubMed  Google Scholar 

  52. Alam, N. A. et al. Genetic and functional analyses of FH mutations in multiple cutaneous and uterine leiomyomatosis, hereditary leiomyomatosis and renal cancer, and fumarate hydratase deficiency. Hum. Mol. Genet. 12, 1241–1252 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Gossage, L., Cartwright, E., Eisen, T. & Bycroft, M. A detailed analysis of von Hippel Lindau (VHL) mutations in sporadic clear cell renal carcinoma (ccRCC), VHL syndrome, and Chuvash polycythaemia. J. Clin. Oncol. 28, e15024–e15024 (2010).

    Article  Google Scholar 

  54. Liang, J. et al. APC polymorphisms and the risk of colorectal neoplasia: a HuGE review and meta-analysis. Am. J. Epidemiol. 177, 1169–1179 (2013).

    Article  PubMed  Google Scholar 

  55. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Statist. Soc. B Methodol. 57, 289–300 (1995).

    Google Scholar 

  56. Bielski, C. M. et al. Widespread selection for oncogenic mutant allele imbalance in cancer. Cancer Cell 34, 852–862.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zehir, A. et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 23, 703–713 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Chang, M. T. et al. Accelerating discovery of functional mutant alleles in cancer. Cancer Discov. 8, 174–183 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Vogelstein, B. et al. Cancer genome landscapes. Science 339, 1546–1558 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank our patients and their families for participating in this study. We thank the members of the Molecular Diagnostics Service in the Department of Pathology, the Marie-Josée and Henry R. Kravis Center for Molecular Oncology, the Robert and Kate Niehaus Center for Inherited Cancer Genomics, and the Berger and Taylor labs for discussions and support. This work was supported by National Institutes of Health awards (nos. P30 CA008748 and R01 CA227534 to M.F.B.), the Breast Cancer Research Foundation (to M.E.R. and K.O.), the Fund for Innovation in Cancer Informatics (to J.G.) and Cycle for Survival.

Author information

Authors and Affiliations

Authors

Contributions

P.S., C.B., M.F.B. and B.S.T. designed the study. P.S., C.B., P.J., S.S.C., A.L.R., A.V.P., C.M.B., M.F.B. and B.S.T. designed and performed data analysis. C.F., A.S., G.J., M.P., J.H., N.S., R.C., J. Galle, A.Z., M.L., D.M.H., D.B.S. and M.F.B. assisted with prospective genomic and clinical data collection and sample annotation. P.S., C.B., Y.K., S.M., J.V., K.A.C., M.I.C., M.F.W., D.M., O.C.B., L.Z., K.O., M.E.R. and Z.K.S. assisted with germline variant pathogenicity and penetrance annotation. S.O.S., I.D., X.L., J. Gao and N.S. assisted in the development of the SignalDB portal. J.S. assisted with pathology. P.S., C.B., M.F.B. and B.S.T. wrote the manuscript with input from all authors. M.F.B. and B.S.T. contributed equally as senior authors.

Corresponding author

Correspondence to Michael F. Berger.

Ethics declarations

Competing interests

K.A.C. reports advisory or consulting activities with AstraZeneca, MSD Ireland and GSK Ireland, and receives honoraria from Pfizer. M.I.C. has had an advisory role with Pfizer. D.M.H. received personal fees from Chugai Pharma, Boehringer Ingelheim, AstraZeneca, Pfizer, Bayer, Debiopharm Group and Genentech, and grants from AstraZeneca, Puma Biotechnology, Loxo Oncology (now owned by Eli Lilly) and Bayer. L.Z. received honoraria from Future Technology Research LLC, Roche Diagnostics Asia Pacific, BGI and Illumina, and has family members with leadership positions and ownership interests in Decipher Medicine. M.E.R. reports honoraria from Research to Practice, Intellisphere and Physician’s Education Resource; consulting and advisory activities for AstraZeneca, Daiichi-Sankyo, Epic Science, Merck and Pfizer (all uncompensated), and Change Healthcare; institutional research funding from AbbVie, AstraZeneca, Merck and Pfizer; and editorial services for AstraZeneca and Pfizer. D.B.S. has consulted with and received honoraria from Pfizer, Loxo/Lilly Oncology, Illumina, Vividion Therapeutics, Scorpion Therapeutics, Fore Biotherapeutics and BioBridge Pharma. Z.K.S. has an immediate family member who serves as a consultant in Ophthalmology for Alcon, Adverum, Gyroscope Therapeutics Ltd, Neurogene and RegenexBio, outside the submitted work. M.F.B. reports receiving research funding from Illumina and Grail and advisory board activities for Roche. B.S.T. reports advisory board activities for Boehringer Ingelheim and Loxo Oncology at Lilly, and research support from Genentech. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Stephen J. Chanock, Clare Turnbull and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Variant discovery and pathogenicity classification.

a) Schematic of the workflow for the germline variant discovery pipeline. b) Contribution of most important features used in classification of pathogenicity. c) Evidence for pathogenicity in classifier-based pathogenicity calls. Proportion of variant calls predicted as pathogenic (first four columns) or benign (fifth column) that exhibited orthogonal evidence of pathogenicity from ClinVar (April, 2020), by medical geneticist review, or as truncating mutations in tumor suppressor genes (excluding last exon or within 50 amino acids at the C-terminus end of the protein).

Extended Data Fig. 2 Ancestry inference.

a) The inference of ancestry from polymorphic SNPs with sufficient coverage in the MSK-IMPACT assay (see Supplementary Note). b) Breakdown of pathogenic variants by ancestry subpopulation. EUR, European; ASJ, Ashkenazi Jewish; AFR, African/African American; ASN, East/South Asian; OTH, Other.

Extended Data Fig. 3 Mechanisms of biallelic inactivation.

Different classes of somatic alterations leading to biallelic loss in carriers of pathogenic variants are shown by gene. LOH, Copy number loss of heterozygosity.

Extended Data Fig. 4 Biallelic inactivation via epigenetic means in TCGA germline carriers.

a) The number of carriers of one of 310 pathogenic germline mutations in 17 high penetrance genes for which biallelic inactivation was apparent by promoter methylation (dark blue; n = 10 total, 3.2% of such patients). Data suggests that heterozygous carriers do not acquire biallelic inactivation via epigenetic silencing of the remaining allele in appreciable numbers as an alternative mechanism to LOH. b) Germline mutations and promoter methylation combined for BRCA1 in ovarian cancers and MLH1 in colorectal cancers indicate that they arise mutually exclusively in affected cancers. All data were acquired from the PanCancerAtlas of The Cancer Genome Atlas project (see Supplementary Note).

Extended Data Fig. 5 Tumor-specific zygosity inference by various classes of variants and specimen types.

a) The rate of biallelic inactivation in the tumors for pathogenic variants as well as multiple classes of non-pathogenic variants including all variants of unknown significance (VUS, as in Fig. 1b), all common variants (MAF > 5%), and all variants annotated as benign in ClinVar. b) The rates of biallelic inactivation in primary and metastasis samples compared with those in benign variants in the corresponding specimen types (primary or metastasis). c) By penetrance level and specimen type (primary or metastasis), the rate of somatic biallelic inactivation of pathogenic variants in cancer types that are either associated or not with increased prevalence in carriers. In gray are benign germline variants in the same genes and cancer types. Points represent biallelic rates for high (n = 714), moderate (n = 354) and low/uncertain (n = 1,353) pathogenic variants. Error bars are 95% binomial CIs. d) As in Main Text Fig. 2c, the rate of somatic biallelic inactivation of germline pathogenic variants in high penetrance genes by association with increased prevalence. Not shown here are those genes with no association to any cancer type or those genes with fewer than five pathogenic variants. Shown are the fraction with somatic biallelic inactivation among carriers of pathogenic variants (red) and benign variants (gray) within the same gene. Error bars are 95% binomial CIs.

Extended Data Fig. 6 Somatic mutations in APC I1307K carriers.

APC I1307K, classified as low penetrance (see Supplementary Note), is a T > A polymorphism that creates a hypermutable tract of eight adenines that increases the propensity for polymerase slippage leading to an additional insertion of adenine, which generates a frameshift. In our cohort, this somatic APC I1307fs* frameshift mutation that results from the aforementioned polymerase slippage on the allele carrying the germline I1307K variant occurred only in colorectal cancers. Seven of these eight colorectal cancers harbored a second somatic mutation leading to biallelic inactivation, which reaffirmed the ‘three-hit’ model for this variant.

Extended Data Fig. 7 Age of onset in germline carriers by association with cancer type.

Age of cancer diagnosis is shown for carriers of high penetrance pathogenic germline variants stratified by association with cancer type and zygosity. Data shown for 10,076 germline WT patients, 330 carriers in associated lineages (276 with biallelic loss) and 176 carriers in non-associated lineages (67 with biallelic loss). Linear regression adjusting for cancer type, specimen type (primary vs. metastasis), genomic instability and sex. For the boxplots, the center red line is the median, the lower and the upper hinges represent the first and third quartiles for the ages of onset. The upper and lower whiskers extend up to 1.5 * IQR (interquartile range) above and below the upper and lower hinges, respectively.

Extended Data Fig. 8 Mutational signatures associated with germline alleles.

Spectrum of mutational signatures in germline carriers of pathogenic alleles in the indicated genes. A signature is considered present if 30% or greater of all somatic mutations are attributed to it. Only signatures that are detected in at least 5% of the carriers are shown. In parentheses is the number of carriers of pathogenic alleles that had 10 or more somatic mutations for robust mutational signature inference.

Extended Data Fig. 9 Cancer type-specific differences in somatic alterations in carriers.

A) The gene-specific pattern of somatic alteration differences in GISTs among carriers of high penetrance alleles that are biallelic in the corresponding tumors (left) versus the rest. B) As in panel (A) but for breast cancers harboring canonical PIK3CA or CCND1 alterations (black and gray are carriers of germline alleles that are somatic biallelic or not in the corresponding cancers, respectively). Related to main text Fig. 3.

Extended Data Fig. 10 Somatic phenotypes of MSI-positive tumors in germline MMR carriers.

a) The proportion of germline MMR carriers among patients who presented with Lynch-associated cancers by gene altered, grouped by zygosity and MSI phenotype. b) Tumor mutational burden (TMB) for MSI tumors in carriers of pathogenic germline variants in MLH1, MSH2, MSH6, and PMS2 indicating no significant difference between the mutational burden of MSI tumors in carriers of different MMR gene mutations. For the boxplots, the center line is the median, the lower and the upper hinges represent the first and third quartiles. The upper and lower whiskers extend up to 1.5 * IQR (interquartile range) above and below the upper and lower hinges, respectively. c) MSIsensor score as a function of the proportion of indels among somatic mutations is shown for MSS tumors (black) compared to those germline pathogenic MSH6 (light blue), PMS2 (dark blue), MLH1 (light green), and MSH2 (dark green) carriers indicate gene-specific differences in their somatic mutational phenotype. Specifically, MSH6-mutant patients had a lower intensity of the MSI phenotype as measured by MSIsensor (P = 2.1 × 10-3, Mann Whitney U test) and a lower proportion of somatic indels in affected tumors (P = 3.7 × 10-7, Mann Whitney U test). d) Three distinct classes of somatic mutations (insertions, deletions, and substitutions) in the affected tumors of germline carriers of the indicated MMR genes (same as in panel c) indicates different germline MMR dysfunctions drive mutation class-specific differences in the somatic MSI phenotype. For the boxplots, the center line is the median, the lower and the upper hinges represent the first and third quartiles. The upper and lower whiskers extend up to 1.5 * IQR (interquartile range) above and below the upper and lower hinges, respectively. e) Immunogenic burden, determined as the ratio of total number of mutation derived epitopes that are strong binders to the total number of non-synonymous mutations, is shown for MSI tumors harboring pathogenic germline variants in MMR genes along with tumors that are germline wild-type (Non-carrier, including non-MSI tumors) with TMB > 20. Tumors from carriers of germline mutations in MLH1/MSH2 had significantly higher immunogenic burden than those tumors that are carriers of germline mutations in MSH6/PMS2 (P = 1.2 × 10-5, after adjusting for tumor type and sample type). Compared with Non-carriers, carriers of germline mutations in MLH1 and MSH2 had significantly higher immunogenic burden (P = 1.3 × 10-5, P = 8 × 10-4, respectively, Wilcoxon test) while carriers with germline mutations MSH6 and PMS2 did not differ from Non-carriers. For the boxplots, the center line is the median, the lower and the upper hinges represent the first and third quartiles. The upper and lower whiskers extend up to 1.5 * IQR (interquartile range) above and below the upper and lower hinges, respectively.

Supplementary information

Supplementary Information

Supplementary Tables 1–6, Note and Figs. 1–10.

Reporting Summary

Supplementary Tables

Supplementary Table 1 Cancer types in cohort. The number of samples of each cancer type among those studied here along with OncoTree node mapping. Supplementary Table 2 Gene and variant level stratification by penetrance. The penetrance level of genes and alleles utilized here. Supplementary Table 3 Germline pathogenicity by cancer type and penetrance. The percentage of patients with the indicated cancer types that were carriers of germline pathogenic alleles are shown, stratified by penetrance level. Supplementary Table 4 Germline pathogenicity by gene and penetrance. The percentage of patients who were carriers of pathogenic alleles in the indicated genes, stratified by penetrance level. Supplementary Table 5 Gene and cancer type associations. The associations among genes and cancer types for those pathogenic alleles shown by ancestry-corrected association testing in the study cohort or those curated by literature review. Excluded here were those genes and cancer types for which no prior evidence was found and insufficient power was available for testing.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Srinivasan, P., Bandlamudi, C., Jonsson, P. et al. The context-specific role of germline pathogenicity in tumorigenesis. Nat Genet 53, 1577–1585 (2021). https://doi.org/10.1038/s41588-021-00949-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00949-1

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer