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:

Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes

Abstract

We studied the whole-genome point mutation and structural variation patterns of 133 tumors (59 high-grade serous (HGSC), 35 clear cell (CCOC), 29 endometrioid (ENOC), and 10 adult granulosa cell (GCT)) as a substrate for class discovery in ovarian cancer. Ab initio clustering of integrated point mutation and structural variation signatures identified seven subgroups both between and within histotypes. Prevalence of foldback inversions identified a prognostically significant HGSC group associated with inferior survival. This finding was recapitulated in two independent cohorts (n = 576 cases), transcending BRCA1 and BRCA2 mutation and gene expression features of HGSC. CCOC cancers grouped according to APOBEC deamination (26%) and age-related mutational signatures (40%). ENOCs were divided by cases with microsatellite instability (28%), with a distinct mismatch-repair mutation signature. Taken together, our work establishes the potency of the somatic genome, reflective of diverse DNA repair deficiencies, to stratify ovarian cancers into distinct biological strata within the major histotypes.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Integration of genomic features stratifies ovarian cancer histotypes.
Figure 2: Foldback inversion profile stratifies patients with HGSC.
Figure 3: Association between foldback inversions and high-level amplifications, with validation on TCGA data.
Figure 4: Stratification of endometriosis-associated tumors.
Figure 5: Overview of ovarian tumor subgroupings by the genomic consequences of aberrant DNA repair.

Similar content being viewed by others

References

  1. Köbel, M. et al. Ovarian carcinoma subtypes are different diseases: implications for biomarker studies. PLoS Med. 5, e232 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Vaughan, S. et al. Rethinking ovarian cancer: recommendations for improving outcomes. Nat. Rev. Cancer 11, 719–725 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Risch, H.A. et al. Population BRCA1 and BRCA2 mutation frequencies and cancer penetrances: a kin-cohort study in Ontario, Canada. J. Natl. Cancer Inst. 98, 1694–1706 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Alsop, K. et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation–positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J. Clin. Oncol. 30, 2654–2663 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Berns, E.M. & Bowtell, D.D. The changing view of high-grade serous ovarian cancer. Cancer Res. 72, 2701–2704 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. Anglesio, M.S., Carey, M.S., Köbel, M., Mackay, H. & Huntsman, D.G. Clear cell carcinoma of the ovary: a report from the first Ovarian Clear Cell Symposium, June 24th, 2010. Gynecol. Oncol. 121, 407–415 (2011).

    Article  PubMed  Google Scholar 

  7. Munksgaard, P.S. & Blaakaer, J. The association between endometriosis and ovarian cancer: a review of histological, genetic and molecular alterations. Gynecol. Oncol. 124, 164–169 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Ahmed, A.A. et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J. Pathol. 221, 49–56 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  10. Wiegand, K.C. et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N. Engl. J. Med. 363, 1532–1543 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Jones, S. et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330, 228–231 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Obata, K. et al. Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res. 58, 2095–2097 (1998).

    CAS  PubMed  Google Scholar 

  13. Wu, R., Zhai, Y., Fearon, E.R. & Cho, K.R. Diverse mechanisms of β-catenin deregulation in ovarian endometrioid adenocarcinomas. Cancer Res. 61, 8247–8255 (2001).

    CAS  PubMed  Google Scholar 

  14. Campbell, I.G. et al. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res. 64, 7678–7681 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Kuo, K.-T. et al. Frequent activating mutations of PIK3CA in ovarian clear cell carcinoma. Am. J. Pathol. 174, 1597–1601 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kurman, R.J. & Shih, IeM. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer—shifting the paradigm. Hum. Pathol. 42, 918–931 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. McConechy, M.K. et al. Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas. J. Pathol. 223, 567–573 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Nissenblatt, M. Endometriosis-associated ovarian carcinomas. N. Engl. J. Med. 364, 482–483, author reply 484–485 (2011).

    Article  CAS  PubMed  Google Scholar 

  19. Wu, R.-C. et al. Frequent somatic mutations of the telomerase reverse transcriptase promoter in ovarian clear cell carcinoma but not in other major types of gynaecological malignancy. J. Pathol. 232, 473–481 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Niskakoski, A. et al. Distinct molecular profiles in Lynch syndrome–associated and sporadic ovarian carcinomas. Int. J. Cancer 133, 2596–2608 (2013).

    CAS  PubMed  Google Scholar 

  21. Shah, S.P. et al. Mutation of FOXL2 in granulosa-cell tumors of the ovary. N. Engl. J. Med. 360, 2719–2729 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Piccart, M.J. et al. Randomized intergroup trial of cisplatin–paclitaxel versus cisplatin–cyclophosphamide in women with advanced epithelial ovarian cancer: three-year results. J. Natl. Cancer Inst. 92, 699–708 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Rauh-Hain, J.A. & Penson, R.T. Potential benefit of sunitinib in recurrent and refractory ovarian clear cell adenocarcinoma. Int. J. Gynecol. Cancer 18, 934–936 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. McAlpine, J.N. et al. HER2 overexpression and amplification is present in a subset of ovarian mucinous carcinomas and can be targeted with trastuzumab therapy. BMC Cancer 9, 433 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Anglesio, M.S. et al. IL6–STAT3–HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Clin. Cancer Res. 17, 2538–2548 (2011).

    Article  CAS  PubMed  Google Scholar 

  26. Farley, J.H., Gibson, S.J. & Monk, B.J. American Society of Clinical Oncology 2012 annual meeting update: summary of selected gynecologic cancer abstracts. Gynecol. Oncol. 126, 319–324 (2012).

    Article  PubMed  Google Scholar 

  27. Anglesio, M.S. et al. Molecular characterization of mucinous ovarian tumours supports a stratified treatment approach with HER2 targeting in 19% of carcinomas. J. Pathol. 229, 111–120 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Ledermann, J. et al. Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N. Engl. J. Med. 366, 1382–1392 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Ledermann, J. et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol. 15, 852–861 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Mirza, M.R. et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N. Engl. J. Med. 375, 2154–2164 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Swisher, E.M. et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 18, 75–87 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Campbell, P.J. et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Sudmant, P.H. et al. An integrated map of structural variation in 2,504 human genomes. Nature 526, 75–81 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ng, C.K.Y. et al. The role of tandem duplicator phenotype in tumour evolution in high-grade serous ovarian cancer. J. Pathol. 226, 703–712 (2012).

    Article  CAS  PubMed  Google Scholar 

  37. Sasaki, S. et al. Molecular processes of chromosome 9p21 deletions in human cancers. Oncogene 22, 3792–3798 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Yang, L. et al. Diverse mechanisms of somatic structural variations in human cancer genomes. Cell 153, 919–929 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hermetz, K.E. et al. Large inverted duplications in the human genome form via a fold-back mechanism. PLoS Genet. 10, e1004139 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Patch, A.-M. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 521, 489–494 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Tothill, R.W. et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin. Cancer Res. 14, 5198–5208 (2008).

    Article  CAS  PubMed  Google Scholar 

  42. Ottaviani, D., LeCain, M. & Sheer, D. The role of microhomology in genomic structural variation. Trends Genet. 30, 85–94 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Sfeir, A. & Symington, L.S. Microhomology-mediated end joining: a back-up survival mechanism or dedicated pathway? Trends Biochem. Sci. 40, 701–714 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Le'tourneau, I.J. et al. Derivation and characterization of matched cell lines from primary and recurrent serous ovarian cancer. BMC Cancer 12, 1 (2012).

    Article  Google Scholar 

  45. Bashashati, A. et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 231, 21–34 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Schwarz, R.F. et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med. 12, e1001789 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Mateos-Gomez, P.A. et al. Mammalian polymerase θ promotes alternative NHEJ and suppresses recombination. Nature 518, 254–257 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Ceccaldi, R. et al. Homologous-recombination-deficient tumours are dependent on Polθ-mediated repair. Nature 518, 258–262 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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 

  50. Swanton, C., McGranahan, N., Starrett, G.J. & Harris, R.S. APOBEC enzymes: mutagenic fuel for cancer evolution and heterogeneity. Cancer Discov. 5, 704–712 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Taghiyar, M.J. et al. Kronos: a workflow assembler for genome analytics and informatics. Preprint at bioRxiv http://dx.doi.org/10.1101/040352 (2016).

Download references

Acknowledgements

We wish to acknowledge generous long-term funding support from the BC Cancer Foundation, supporting the research program of S.P.S. and OvCaRe. The authors graciously thank the Gray Family Ovarian Clear Cell Carcinoma Research Resource, which provided funding critical to this project. Additional funding was provided by a Terry Fox Research Institute New Investigator grant to S.P.S. and a Canadian Cancer Society Research Institute Impact grant to D.G.H. and S.P.S. The CRCHUM Ovarian Cancer Tumour Bank was supported by the Réseau de Recherche sur le Cancer, Fonds de Recherche Québec Santé, affiliated with the Canadian Tumour Repository Network. S.P.S. is a Michael Smith Foundation for Health Research (MSFHR) scholar and holds a Canadian Institutes for Health Research (CIHR) Foundation grant. S.P.S. and D.G.H. hold Canada Research Chairs. Y.K.W. is an MSFHR postdoctoral fellow. Support for the development of software used in this study was provided by the Genome Canada and Genome British Columbia Bioinformatics and Computational Biology program. Finally, the authors wish to acknowledge the funding support to S.P.S. from the Discovery Frontiers: Advancing Big Data Science in Genomics Research program (grant RGPGR/448167-2013, 'The Cancer Genome Collaboratory'), which is jointly funded by the Natural Sciences and Engineering Research Council of Canada, the Canadian Institutes of Health Research, Genome Canada, and the Canada Foundation for Innovation and with in-kind support from the Ontario Research Fund of the Ministry of Research, Innovation, and Science.

Author information

Authors and Affiliations

Authors

Contributions

Y.K.W. and A.B. were the research project leaders and led and designed all data analysis. M.S.A. performed molecular subtype classification of HGSC samples. M.S.A., D.R.C., and H.M.H. collected CCOC, ENOC, and GCT samples and conducted experiments. D.S.G., G.H., A.M., D.L., M.R.A., A.W.Z., K.S., and C.S. performed data analysis and bioinformatics. L.M.P., J.S., M.K.M., and A.W. performed sample preparation and validation experiments. A.O., S.Y., N.Y., M.S., A.T., D.P., M.d.L., and H.F. were responsible for specimens and clinical data from Tokyo and Montreal. R.M., A.J.M., and M.A.M. performed library construction and genome sequencing. C.B.G., H.M.H., A.N.K., and H.L.-C. reviewed all specimens for histological and molecular pathology. J.N.M. and A.-M.M.-M. performed cohort design, HGSC sample selection, and tumor banking. S.A. oversaw experimental design and genome sequencing for validation. Y.K.W., A.B., and S.P.S. wrote the manuscript. D.G.H. conceived the project, provided oversight, and edited the manuscript. S.P.S. conceived and oversaw the project and is the senior responsible author.

Corresponding authors

Correspondence to David G Huntsman or Sohrab P Shah.

Ethics declarations

Competing interests

S.P.S., D.G.H., and S.A. are founders and shareholders of Contextual Genomics, Inc.

Integrated supplementary information

Supplementary Figure 1 Patterns of somatically acquired genomic variants in GCT, CCOC, ENOC, and HGSC ovarian cancers.

(a) Top, mutation load showing the total number of mutations (y axis; log10 scale) for each tumor (x axis). Samples are sorted in descending order based on the total number of mutations. Second panel, contributions of the six identified mutation signatures per sample. Six mutation signatures were extracted from the trinucleotide substitutions of 133 tumor genomes: S.APOBEC, signature similar to the COSMIC APOBEC signature (COSMIC signature 13); S.POLE, a mimic of COSMIC signature 10 related to altered activity of the error-prone polymerase POLE; S.AGE, the age-related signature (COSMIC signature 1) that has been known to correlate with age at cancer diagnosis; S.BC, closely matched with the pattern of COSMIC signature 8 previously found in breast cancer and medulloblastoma; S.MMR, matched to COSMIC signature 6 associated with defective mismatch repair; S.HRD, associated with COSMIC signature 3 representing deficiency in homologous recombination DNA repair. (b) Proportion of the genome harboring high-level copy number amplifications (AMP; top), dominant loss of heterozygosity (LOH; second panel), and copy number loss (LOSS; bottom) per sample. (c) Total number of rearrangements (top; y axis, log10 scale) and the proportion of rearrangement types (second panel) observed in each sample. Balanced, balanced rearrangements; Deletion, deletion rearrangements; Duplication, tandem duplications; Foldback, foldback inversions; Unbalanced, unbalanced rearrangements; Inversion, inversion rearrangements.

Supplementary Figure 2 Substitution mutational signatures.

(a) Top, total number of SNVs (y axis; log10 scale) for each tumor (x axis). Second panel, proportion of six base substitution patterns per sample. Samples are sorted in descending order according to the total number of mutations. (b) Residual sum of squares (RSS; top) and explained variance (second panel) for the number of signatures from 2 to 12, each with 200 replicates. (c) Six inferred mutational signature profiles. S.APOBEC, signature similar to the COSMIC APOBEC signature (COSMIC signature 13); S.POLE, a mimic of COSMIC signature 10 related to altered activity of the error-prone polymerase POLE; S.AGE, the age-related signature (COSMIC signature 1) that has been known to correlate with age at cancer diagnosis; S.BC, closely matched with the pattern of COSMIC signature 8 previously found in breast cancer; S.MMR, matched to COSMIC signature 6 associated with defective mismatch repair; S.HRD, associated with COSMIC signature.3 representing deficiency in homologous recombination DNA repair.

Supplementary Figure 3 Genomic feature descriptions.

Description of the 20 genomic features used in the integrative clustering of patients with ovarian cancer, including 6 mutation signatures (S.APOBEC, S.POLE, S.AGE, S.BC, S.MMR, and S.HRD); 6 rearrangement types and 1 homology length (Foldback.Inversion, Inversion, Tandem.Duplication, Deletion.Rearrangement, Balanced.Rearrangement, Unbalanced.Rearrangement, and Homology>=5bp); 3 copy number aberrations (CN.Amplification, CN.Loss, and CN.LOH); and 4 mutation variant types (Nonsynonymous, Splicesite, Stop.Lost/Gained, and Frameshift).

Supplementary Figure 4 Integration of genomic features stratifies patients with ovarian cancer.

(a) Hierarchical clustering of 133 patients with ovarian cancer (columns) by integrating genomic features including point mutation, copy number, and structural rearrangement profiles identifies seven major genomic subgroups. Scaled values of genomic features (rows in the top panel) are shown in a heat map with a dendrogram of the hierarchical cluster analysis. The color-coding reflects the scaled value of genomic features obtained by subtracting the value of each feature from its mean and dividing the value by its standard deviation. Mutation status (presence, gray; absence, white; rows for bottom panel) for the significantly mutated genes (MutSigCV q < 0.01) and DNA repair genes across patients is displayed. Histotype is included as an annotation row (red, HGS; blue, COCC; green, ENOC; purple, GCT). (b) Comparison of the estimated cellularities of the subgroups of each histotype, where no significant differences were observed. The cellularity of each sample was estimated using Titan. Student’s t test was performed, and the corresponding P value is annotated on top of the box plots for each histotype. (c) Determining the number of clusters by the ‘elbow’ rule. The plot shows the explained variance (EV; y axis) computed as a function of the number of clusters (x axis) generated from hierarchical clustering. Given the threshold of EV (at 0.45; horizontal dashed line) and its increment threshold of 0.05, the optimal number of clusters (k = 7) was identified (vertical dashed line). (d) Mutation load in the HGSC subgroups. The mutation load for the HGSC samples in the H-HRD subgroup, on average, was higher than in the H-FBI subgroup (Mann–Whitney–Wilcoxon test, P < 0.001). (e) Focal amplifications (red) and deletions (blue) in the CCOC (C-APOBEC and C-AGE) subgroups. (f) Focal amplifications (red) and deletions (blue) in E-MSI and MSS ENOC samples.

Supplementary Figure 5 Integration/clustering of genomic features/cases in the HGSC cohort only.

(a) Hierarchical clustering of 59 patients with HGSC by integrating genomic variant profiles highlights two major genomic subgroups, H-FBI (n = 22) and H-HRD (n = 37), of HGSC tumors. The plot shows the contribution of genomic features (scaled value; top heat map) with a dendrogram illustrating hierarchical clustering. The mutation status (presence, gray; absence, white) for the significantly mutated genes (MutSigCV q < 0.01) and DNA repair genes across patients is shown in the second panel. Histotype and two subgroups are included as annotation rows (red, HGSC; dark green, H-FBI subgroup; dark orange, H-HRD subgroup). (b) Importance of genomic features segregating the HGSC subgroups of H-FBI (n = 22) and H-HRD (n = 37). Left, genomic features (y axis) are sorted in descending order of the average Gini score (x axis), reflecting the importance of features in stratifying the two subgroups of HGSC tumors. Right, box plot showing the distribution of the top six genomic features contributing to the differences between H-HRD and H-FBI. The y axis shows the value of genomic features. (c,d) Kaplan–Meier plots showing significant differences in overall survival (c) and progression-free survival (d) between the HGSC subgroups H-HRD and H-FBI (log-rank test, P = 0.0083 and 0.0108), in which samples enriched in foldback inversions (H-FBI) had poor survival outcomes.

Supplementary Figure 6 Integration/clustering of genomic features/cases in patients with endometrioisis-associated cancers only.

(a) Hierarchical clustering of 35 patients with CCOC and 29 patients with ENOC by integrating genomic variant profiles highlights six major genomic subgroups. The plot shows the contribution of genomic features (scaled value; top heat map) with a dendrogram illustrating hierarchical clustering. The mutation status (presence, gray; absence, white) for the significantly mutated genes (MutSigCV q < 0.01) and DNA repair genes across patients is displayed. (b) Contribution of genomic subgroup memberships in ENOC and CCOC. The number (n) and proportion (%) of samples from each subgroup are shown. (c) Importance of genomic features segregating the C-APOBEC (n = 9) and C-AGE (n = 15) subgroups of CCOC tumors. Left, genomic features (y axis) are sorted in descending order of the average Gini score (x axis), reflecting the importance of features in stratifying the two subgroups of CCOC tumors. Right, box plot showing the distribution of the top six genomic features contributing to the differences between C-APOBEC and C-AGE. The y axis shows the value of genomic features. (d) Importance of genomic features segregating the E-MSI (n = 8) and MSS (n = 20) subgroups of ENOC tumors. Left, genomic features (y axis) are sorted in descending order of the average Gini score (x axis), reflecting the importance of features in stratifying MSI and E-MSS ENOC tumors. Right, box plot showing the distribution of the top six genomic features contributing to the differences between the E-MSI and MSS subgroups. The y axis shows the value of genomic features. (e) Box plots showing the distribution of immunogenic epitope counts in the MSS and E-MSI ENOC subgroups.

Supplementary Figure 7 Homozygous deletions identified from HGSC tumors in PTEN and RB1.

(a,b) Examples of homozygous deletion in PTEN (a) and RB1 (b). In each example, the following are shown: a chromosome ideogram highlighting the region of interest (top); a log-ratio plot overlaying rearrangement events (if present, shown with arcs) on copy number aberration segments (middle); an allelic ratio plot showing the corresponding LOH profile in each region (bottom). ALOH, amplified LOH; HOMD, homozygous deletion; NLOH, neutral LOH; Deletion, deletion rearrangement.

Supplementary Figure 8 HGSC tumors stratified by foldback-inversion profile. 

(a) HGSC cases could be stratified into two subgroups based on the proportion of foldback inversions, in which cases with a higher proportion of foldback inversions (with reference to the median) referred to as the High FBI group had statistically significant inferior overall and progression-free survival outcomes (log-rank test, P = 0.0187 and 0.0286) as compared to cases with a low proportion of foldback inversions (Low FBI group). (b) Distribution of the break distance for foldback inversions in our HGSC cohort. (cf) Two examples of foldback inversions at chromosome 8. In c and e, the genomic locus of the events is illustrated. A red bar marked on an ideogram of the chromosome 8 q arm shows where the events occurred (top). Two foldback inversions are illustrated: SV1, on the forward strand (arrows pointing to the right in orange; second panel) and SV2, on the reverse strand (backward arrows pointing to the left in grey; third panel). Coverage depth and reads (fourth and fifth panels) covering the breakpoints of foldback inversions and the locations of breakpoints on the genomic scale (bottom) are shown. In d and f, two foldback inversions are shown schematically at nucleotide sequence level. Annotated red on the genomic scale shows the breakpoints of the forward strand and the reverse strand sequences. In d, two foldback inversions co-occurred within 1 kb. Left, the reverse strand inverted and was fused to the forward strand by a 4-bp homology sequence, CTTT (highlighted in green). Right, the forward strand inverted and was fused to the reverse strand by a 12-bp homology sequence, TTCACATGTGAA (highlighted in green). In f, two foldback inversions co-occurred within 2 kb. Left, the reverse strand inverted and was fused to the forward strand by a 4-bp homology sequence, GAGC (highlighted in green). Right, the forward strand inverted and was fused to the forward strand by a 12-bp homology sequence, AGAGTATACTCT (highlighted in green).

Supplementary Figure 9 Co-occurrence of foldback inversions and focal high-level amplifications (HLAMPs) in HGSC samples.

Examples of focal HLAMPs colocalized with foldback inversions in CCNE1 and KRAS in our discovery HGSC cohort and in CCNE1 and MYC in the ICGC HGSC cohort. In each example, the following are shown: a chromosome ideogram highlighting the region of interest (top); a log-ratio plot overlaying rearrangement events (shown with arcs) on copy number (CN) aberration segments (middle); an allelic ratio plot showing the corresponding LOH profile in each region (bottom). ALOH, amplified LOH; ASCNA, allele-specific copy number amplification; BCNA, balanced copy number amplification; GAIN, copy number gain; HET, diploid heterozygous; NLOH, neutral LOH.

Supplementary Figure 10 High-level amplification–associated foldback inversions (HLAMP-FBIs) in HGSC cell lines.

(a) The proportion of HLAMP-FBI for the primary (TOV1369) and relapse (OV1369(R2) cell lines) (red dotted lines) superimposed on the distribution of HLAMP-FBI from the H-HRD (blue) and H-FBI (green) subgroups. (b) Examples of HLAMP-associated foldback inversions present in the relapse cell line (OV1369(R2)) but absent in the primary-tumor-derived cell line (TOV1369) from the same patient. In each example, the following are shown: a chromosome ideogram highlighting the region of interest (top); log-ratio plots overlaying rearrangement events (shown with arcs) on copy number (CN) aberration segments in the primary cell line (middle) and relapse cell line (bottom). HOMD, homozygous deletion; DLOH, deletion LOH; HET, diploid heterozygous; GAIN, CN gain; AMP, copy number amplification; HLAMP, high-level amplification.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Note (PDF 3715 kb)

Supplementary Table 1

Clinical information of all samples in the study. (XLSX 67 kb)

Supplementary Table 2

Targeted deep sequencing analysis for validating case-specific SNV calls. (XLS 3492 kb)

Supplementary Table 3

PCR validation experiment on the selected breakpoints identified from sample DAH208 to determine breakpoint filtering criteria for destruct predictions. (XLSX 50 kb)

Supplementary Table 4

Sample, sequencing, and variant summary information. (XLSX 77 kb)

Supplementary Table 5

Genomic feature matrix and sample–gene variant table. (XLSX 100 kb)

Supplementary Table 6

Significantly mutated genes (SMGs) identified from all ovarian samples inferred by MutSigCV. (XLSX 1782 kb)

Supplementary Table 7

Feature ranking by shrinkage discriminant analysis. (XLSX 59 kb)

Supplementary Table 8

Feature importance table. (XLSX 18 kb)

Supplementary Table 9

Mutations found in the non-BRCA1/2 BROCA genes in the H-HRD and H-FBI subgroups (XLSX 19 kb)

Supplementary Table 10

Focal amplifications and deletions. (XLSX 170 kb)

Supplementary Table 11

ICGC HGSC cohort stratified by foldback-inversion profile. (XLSX 71 kb)

Supplementary Table 12

Comparisons of copy number gains/amplifications associated with rearrangement events. (XLSX 67 kb)

Supplementary Table 13

TCGA HGSC cohort stratified by FBI-AMP profile. (XLSX 64 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Bashashati, A., Anglesio, M. et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nat Genet 49, 856–865 (2017). https://doi.org/10.1038/ng.3849

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3849

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing