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:

Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer

Abstract

High-grade serous ovarian cancer (HGSC) is frequently characterized by homologous recombination (HR) DNA repair deficiency and, while most such tumors are sensitive to initial treatment, acquired resistance is common. We undertook a multiomics approach to interrogate molecular diversity in end-stage disease, using multiple autopsy samples collected from 15 women with HR-deficient HGSC. Patients had polyclonal disease, and several resistance mechanisms were identified within most patients, including reversion mutations and HR restoration by other means. We also observed frequent whole-genome duplication and global changes in immune composition with evidence of immune escape. This analysis highlights diverse evolutionary changes within HGSC that evade therapy and ultimately overwhelm individual patients.

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: End-stage HGSC cohort, samples and experimental modalities.
Fig. 2: Clonal composition of end-stage HGSC.
Fig. 3: Summary of HGSC genomic landscape at autopsy, as assessed by WGS.
Fig. 4: Known resistance mechanisms in HR-deficient HGSC.
Fig. 5: Multisite and multitemporal immune profiling of HR-deficient HGSC.
Fig. 6: Proteomic-level data analysis.
Fig. 7: Summary of evolutionary events and resistance mechanisms detected for each patient.

Similar content being viewed by others

Data availability

ICGC dataset: previously published WGS and RNA-seq data generated as part of the ICGC Ovarian Cancer project are available from the European Genome-phenome Archive (EGA) repository (https://ega-archive.org) as a single bam file for each sample type (tumor/normal), under accession code EGAD00001000877. Due to the sensitive nature of these patient datasets, access is subject to approval from the ICGC Data Access Compliance Office (https://docs.icgc.org/download/data-access/), an independent body that authorizes controlled access to ICGC sequencing data. ICGC SNP array and methylation datasets have been deposited in Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/) under accession code GSE65821, without access restrictions. ICGC gene count-level transcriptomic data have been deposited in the GEO under accession code GSE209964. CASCADE dataset: WGS, RNA-seq and SNP array data from participants in a rapid research autopsy generated as part of this CASCADE study have been deposited in the EGA repository under accession code EGAS00001006789. WGS (no. EGAD00001009746) and RNA-seq data (no. EGAD00010002398) are available as raw FASTQ files for each sample type (tumor/normal and tumor, respectively), and SNP array data are available as raw signal intensity files in text format for each sample type (tumor/normal, no. EGAD00010002398). Targeted sequencing data for the same participants are available as raw FASTQ files for both tumor and normal (no. EGAD00001009747). Due to the sensitive nature of these patient datasets, access can be gained for academic use through application to the independent Data Access Committee. Further information on how to apply for access is available at EGA under accession code EGAS00001006789. Transcriptomic count-level data and methylation data for the same participants has been deposited in the GEO under accession code GSE217672 (RNA) and GSE217673 (methylation), available at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE217674, without access restrictions. Mutational signature reference databases can be accessed via COSMIC (https://cancer.sanger.ac.uk/signatures/) and Signal (https://signal.mutationalsignatures.com/). The LM22 signature matrix used for immune cell deconvolution can be downloaded from https://cibersortx.stanford.edu/. The COSMIC Cancer Gene Census can be accessed from https://cancer.sanger.ac.uk/census. MSigDB hallmark gene sets can be accessed from https://www.gsea-msigdb.org/gsea/msigdb/. Illumina methylation probes that were filtered out due to poor performance (for example, cross-reactive or nonspecific probes) can be found at https://github.com/sirselim/illumina450k_filtering. Germline polymorphic sites for reference and variant allele read counts used in FACETS analysis can be found at ftp://ftp.ncbi.nih.gov/snp/organisms/human_9606_b151_GRCh37p13/VCF/common_all_20180423.vcf.gz. The GTF used for annotation and RNA-seq counts are available at ftp://ftp.ensembl.org/pub/grch37/release-92/. All other data are available within the article and its Supplementary Information files. Multicolor immunofluorescence: final cell counts have been uploaded to Synapse under accession no. syn49448783, and are available publicly at https://www.synapse.org/#!Synapse:syn44043685/files/. Proteomics: MS proteomics data have been deposited at the ProteomeXchange Consortium via the PRIDE partner repository, with dataset identifier PXD030034. Detailed methods of data processing from raw files are available in the Supplementary Note. Source data are provided with this paper.

Code availability

R code for all genomics and mcIF analyses and figure generation has been deposited in Synapse (no. syn44044508), available publicly. No custom code was generated for this study.

References

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

    Article  Google Scholar 

  2. Masoodi, T. et al. Genetic heterogeneity and evolutionary history of high-grade ovarian carcinoma and matched distant metastases. Br. J. Cancer 122, 1219–1230 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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).

  4. Priestley, P. et al. Pan-cancer whole-genome analyses of metastatic solid tumours. Nature 575, 210–216 (2019).

  5. Alsop, K., Fereday, S. & Meldrum, C. 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  PubMed  PubMed Central  Google Scholar 

  6. Nguyen, L., Martens, J. W., Van Hoeck, A. & Cuppen, E. Pan-cancer landscape of homologous recombination deficiency. Nat. Commun. 11, 5584 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Pennington, K. P. et al. Germline and somatic mutations in homologous recombination genes predict platinum response and survival in ovarian, Fallopian tube, and peritoneal carcinomas. Clin. Cancer Res. 20, 764–775 (2014).

  8. Song, H. et al. Contribution of germline mutations in the RAD51B, RAD51C, and RAD51D genes to ovarian cancer in the population. J. Clin. Oncol. 33, 2901–2907 (2015).

  9. Ramus, S. et al. Germline mutations in the BRIP1, BARD1, PALB2, and NBN genes in women with ovarian cancer. J. Natl Cancer Inst. 107, djv214 (2015).

  10. Swisher, E. M. et al. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2). Nat. Commun. 12, 2487 (2021).

  11. Alsop, K. et al. A community-based model of rapid autopsy in end-stage cancer patients. Nat. Biotechnol. 34, 1010–1014 (2016).

  12. Lee, S. et al. Molecular analysis of clinically defined subsets of high-grade serous ovarian cancer. Cell Rep. 31, 107502 (2020).

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

  14. Dentro, S. C. et al. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes. Cell 184, 2239–2254 (2021).

  15. Werner, B., Traulsen, A., Sottoriva, A. & Dingli, D. Detecting truly clonal alterations from multi-region profiling of tumours. Sci. Rep. 7, 44991 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Litchfield, K. et al. Representative sequencing: unbiased sampling of solid tumor tissue. Cell Rep. 31, 107550 (2020).

  17. Gillis, S. & Roth, A. PyClone-VI: scalable inference of clonal population structures using whole genome data. BMC Bioinformatics 21, 571 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Gundem, G. et al. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).

  19. Greaves, M. Evolutionary determinants of cancer. Cancer Discov. 5, 806–820 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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).

  21. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

  22. López, S. et al. Interplay between whole-genome doubling and the accumulation of deleterious alterations in cancer evolution. Nat. Genet. 52, 283–293 (2020).

  23. Berenjeno, I. M. et al. Oncogenic PIK3CA induces centrosome amplification and tolerance to genome doubling. Nat. Commun. 8, 1773 (2017).

  24. Lin, K. K. et al. BRCA reversion mutations in circulating tumor DNA predict primary and acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 9, 210–219 (2019).

  25. Christie, E. L. et al. Reversion of BRCA1/2 germline mutations detected in circulating tumor DNA from patients with high-grade serous ovarian cancer. J. Clin. Oncol. 35, 1274–1280 (2017).

  26. Etemadmoghadam, D. et al. Synthetic lethality between CCNE1 amplification and loss of BRCA1. Proc. Natl Acad. Sci. USA 110, 19489–19494 (2013).

  27. Aziz, D. et al. 19q12 amplified and non-amplified subsets of high grade serous ovarian cancer with overexpression of cyclin E1 differ in their molecular drivers and clinical outcomes. Gynecol. Oncol. 151, 327–336 (2018).

  28. Ciriello, G., Cerami, E., Sander, C. & Schultz, N. Mutual exclusivity analysis identifies oncogenic network modules. Genome Res. 22, 398–406 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Norquist, B. et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. J. Clin. Oncol. 29, 3008–3015 (2011).

  30. Bunting, S. F. et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell 141, 243–254 (2010).

  31. Noordermeer, S. M. et al. The shieldin complex mediates 53BP1-dependent DNA repair. Nature 560, 117–121 (2018).

  32. Gallego Romero, I., Pai, A. A., Tung, J. & Gilad, Y. RNA-seq: impact of RNA degradation on transcript quantification. BMC Biol. 12, 42 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Wang, Y. et al. The BRCA1-Δ11q alternative splice isoform bypasses germline mutations and promotes therapeutic resistance to PARP inhibition and cisplatin. Cancer Res. 76, 2778–2790 (2016).

  34. He, Y. J. et al. DYNLL1 binds to MRE11 to limit DNA end resection in BRCA1-deficient cells. Nature 563, 522–526 (2018).

  35. Waks, A. G. et al. Reversion and non-reversion mechanisms of resistance to PARP inhibitor or platinum chemotherapy in BRCA1/2-mutant metastatic breast cancer. Ann. Oncol. 31, 590–598 (2020).

  36. Goode, E. L. et al. Dose-response association of CD8+ tumor-infiltrating lymphocytes and survival time in high-grade serous ovarian cancer. JAMA Oncol. 3, e173290 (2017).

  37. Morse, C. B. et al. Tumor infiltrating lymphocytes and homologous recombination deficiency are independently associated with improved survival in ovarian carcinoma. Gynecol. Oncol. 153, 217–222 (2019).

  38. Sansregret, L., Vanhaesebroeck, B. & Swanton, C. Determinants and clinical implications of chromosomal instability in cancer. Nat. Rev. Clin. Oncol. 15, 139–150 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689 (2018).

  41. Zhang, A. W. et al. Interfaces of malignant and immunologic clonal dynamics in ovarian cancer. Cell 173, 1755–1769 (2018).

  42. Buisson, R. et al. Breast cancer proteins PALB2 and BRCA2 stimulate polymerase η in recombination-associated DNA synthesis at blocked replication forks. Cell Rep. 6, 553–564 (2014).

  43. McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171, 1259–1271 (2017).

  44. Vázquez-García, I. et al. Ovarian cancer mutational processes drive site-specific immune evasion. Nature 612, 778–786 (2022).

  45. De Mattos-Arruda, L. et al. The genomic and immune landscapes of lethal metastatic breast cancer. Cell Rep. 27, 2690–2708 (2019).

  46. Dangaj, D. et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell 35, 885–900 (2019).

  47. Bruand, M. et al. Cell-autonomous inflammation of BRCA1-deficient ovarian cancers drives both tumor-intrinsic immunoreactivity and immune resistance via STING. Cell Rep. 36, 109412 (2021).

  48. Li, M. et al. CCL5 deficiency promotes liver repair by improving inflammation resolution and liver regeneration through M2 macrophage polarization. Cell. Mol. Immunol. 17, 753–764 (2020).

  49. Lan, C. et al. Expression of M2-polarized macrophages is associated with poor prognosis for advanced epithelial ovarian cancer. Technol. Cancer Res. Treat. 12, 259–267 (2013).

  50. Newman, A. M. et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat. Biotechnol. 37, 773–782 (2019).

  51. Castro, A. et al. Elevated neoantigen levels in tumors with somatic mutations in the HLA-A, HLA-B, HLA-C and B2M genes. BMC Med. Genomics 12, 107 (2019).

  52. Sun, J. et al. Immuno-genomic characterisation of high-grade serous ovarian cancer reveals immune evasion mechanisms and identifies an immunological subtype with a favourable prognosis and improved therapeutic efficacy. Br. J. Cancer 126, 1570–1580 (2022).

  53. Hundal, J. et al. pVACtools: a computational toolkit to identify and visualize cancer neoantigens. Cancer Immunol. Res. 8, 409–420 (2020).

  54. Hunt, A. L. et al. Extensive three-dimensional intratumor proteomic heterogeneity revealed by multiregion sampling in high-grade serous ovarian tumor specimens. iScience 24, 102757 (2021).

  55. Benita, Y. et al. An integrative genomics approach identifies Hypoxia Inducible Factor-1 (HIF-1)-target genes that form the core response to hypoxia. Nucleic Acids Res. 37, 4587–4602 (2009).

  56. Bredholt, G. et al. Tumor necrosis is an important hallmark of aggressive endometrial cancer and associates with hypoxia, angiogenesis and inflammation responses. Oncotarget 6, 39676–39691 (2015).

  57. Lin, E. P.-Y. et al. Translating gene signatures into a pathologic feature: tumor necrosis predicts disease relapse in operable and stage I lung adenocarcinoma. JCO Precis. Oncol. 2, 1–13 (2018).

  58. Gonçalves, E. et al. Widespread post-transcriptional attenuation of genomic copy-number variation in cancer. Cell Syst. 5, 386–398 (2017).

  59. Gentric, G. et al. PML-regulated mitochondrial metabolism enhances chemosensitivity in human ovarian cancers. Cell Metab. 29, 156–173. (2019).

  60. Lahiguera, Á. et al. Tumors defective in homologous recombination rely on oxidative metabolism: relevance to treatments with PARP inhibitors. EMBO Mol. Med. 12, e11217 (2020).

  61. Andrabi, S. A. et al. Poly (ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc. Natl Acad. Sci. USA 111, 10209–10214 (2014).

  62. Gallyas, F. Jr & Sumegi, B. Mitochondrial protection by parp inhibition. Int. J. Mol. Sci. 21, 2767 (2020).

    Article  CAS  PubMed  Google Scholar 

  63. Yates, L. R. et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat. Med. 21, 751–759 (2015).

  64. Tobalina, L., Armenia, J., Irving, E., O’Connor, M. & Forment, J. A meta-analysis of reversion mutations in BRCA genes identifies signatures of DNA end-joining repair mechanisms driving therapy resistance. Ann. Oncol. 32, 103–112 (2021).

  65. Clairmont, C. S. et al. TRIP13 regulates DNA repair pathway choice through REV7 conformational change. Nat. Cell Biol. 22, 87–96 (2020).

  66. Dev, H. et al. Shieldin complex promotes DNA end-joining and counters homologous recombination in BRCA1-null cells. Nat. Cell Biol. 20, 954–965 (2018).

  67. Chaudhuri, A. R. et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 535, 382–387 (2016).

  68. Lee, W.-C. et al. Multiomics profiling of primary lung cancers and distant metastases reveals immunosuppression as a common characteristic of tumor cells with metastatic plasticity. Genome Biol. 21, 271 (2020).

  69. Johnson, N. et al. Stabilization of mutant BRCA1 protein confers PARP inhibitor and platinum resistance. Proc. Natl Acad. Sci. USA 110, 17041–17046 (2013).

  70. Tie, J. et al. Circulating tumor DNA analysis guiding adjuvant therapy in stage II colon cancer. N. Engl. J. Med. 386, 2261–2272 (2022).

  71. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020).

  72. Quinton, R. J. et al. Whole genome doubling confers unique genetic vulnerabilities on tumor cells. Nature 590, 492–497 (2021).

  73. Cohen-Sharir, Y. et al. Aneuploidy renders cancer cells vulnerable to mitotic checkpoint inhibition. Nature 590, 486–491 (2021).

  74. Nath, A. et al. Evolution of core archetypal phenotypes in progressive high grade serous ovarian cancer. Nat. Commun. 12, 3039 (2021).

  75. Gaaib, J. N., Nassief, A. F. & Al-Assi, A. Simple salting-out method for genomic DNA extraction from whole blood. Tikrit J. Pure Sci. 16, 9–11 (2011).

  76. Schröder, J., Corbin, V. & Papenfuss, A. T. HYSYS: have you swapped your samples? Bioinformatics 33, 596–598 (2017).

    Article  PubMed  Google Scholar 

  77. Song, S. et al. qpure: A tool to estimate tumor cellularity from genome-wide single-nucleotide polymorphism profiles. PLoS ONE 7, e45835 (2012).

  78. Van Loo, P. et al. Allele-specific copy number analysis of tumors. Proc. Natl Acad. Sci. USA 107, 16910–16915 (2010).

  79. Shen, R. & Seshan, V. E. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 44, e131 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Macintyre, G. et al. Copy number signatures and mutational processes in ovarian carcinoma. Nat. Genet. 50, 1262–1270 (2018).

  81. Chen, X. & Chang, J. T. Planning bioinformatics workflows using an expert system. Bioinformatics 33, 1210–1215 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Bielski, C. M. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat. Genet. 50, 1189–1195 (2018).

  83. Zack, T. I. et al. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).

  84. Liberzon, A. et al. The molecular signatures database hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

  85. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

  86. Bankhead, P. et al. QuPath: Open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

  87. Frangi, A. F. et al. (eds). Proc. MICCAI, 21st International Conference (SpringerLink, 2018).

  88. Fedchenko, N. & Reifenrath, J. Different approaches for interpretation and reporting of immunohistochemistry analysis results in the bone tissue—a review. Diagn. Pathol. 9, 221 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Bakdash, J. Z. & Marusich, L. R. Repeated measures correlation. Front. Psychol. 8, 456 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Brooks, M. E. et al. glmmTMB balances speed and flexibility among packages for zero-inflated generalized linear mixed modeling. https://doi.org/10.32614/RJ-2017-066 (2017)

  91. Hartig, F. DHARMa: residual diagnostics for hierarchical (multi-level/mixed) regression models. https://cran.microsoft.com/snapshot/2021-09-26/web/packages/DHARMa/vignettes/DHARMa.html (2021).

Download references

Acknowledgements

This study was financially supported by grants from the Department of Health and Human Services through the National Health and Medical Research Council of Australia (NHMRC, nos. APP1124309 to E.L.C., APP1161198 to D.D.L.B. and E.L.C., APP1117044 to D.D.L.B., APP1092856 to D.D.L.B., APP1186505 to D.W.G. and APP1189939 to N.L.B.), the US National Cancer Institute U54 program (no. U54CA209978 to A.B., J.T.C. and D.D.L.B.), Victorian Cancer Agency (no. ECSG15012 to E.L.C.), Tour de Cure (no. RSP-274-18/19 to E.L.C.) and Goldman Sachs Gives through the Peter MacCallum Cancer Foundation and Cancer Australia (no. APP1004673 to D.D.L.B.). Part of this study was supported by the US Department of Defense – Uniformed Services University of the Health Sciences (nos. HU0001-16-2-0006 and HU0001-16-2-0014 to G.L.M.). We gratefully acknowledge additional support from M. Rose and the Rose family, The WeirAnderson Foundation, Border Ovarian Cancer Awareness Group, donors to the Garvan Institute of Medical Research’s Ovarian Cancer Research Program, W. Taylor and A. Coombs and family. The CASCADE study was supported by the Peter MacCallum Cancer Centre Foundation, and in kind by the Victorian Institute of Forensic Medicine and Tobin Brothers Funerals. The CASCADE investigators thank the CASCADE Management Committee, all staff at the Victorian Institute of Forensic Medicine, D. Stevens and Tobin Brothers Funerals. We acknowledge and thank the women and their families who generously participated in the CASCADE program making this work possible. We acknowledge the vital role of the Australian Ovarian Cancer Study (AOCS) in this study. AOCS was supported by the US Army Medical Research and Materiel Command (no. DAMD17-01-1-0729 to D.D.L.B.), The Cancer Council Victoria, Queensland Cancer Fund, The Cancer Council New South Wales, The Cancer Council South Australia, The Cancer Foundation of Western Australia and The Cancer Council Tasmania and NHMRC (nos. ID400413 and ID400281 to D.D.L.B.). AOCS acknowledges additional support from Ovarian Cancer Australia and the Peter MacCallum Cancer Centre Foundation. AOCS gratefully acknowledges the cooperation of the participating institutions in Australia, and acknowledges the contribution of the study nurses, research assistants and all clinical and scientific collaborators, in particular L. Bowes, D. Ariyaratne and N. Traficante. We thank E. Niedermayr, S. Guo, all the kConFab research nurses and staff, the heads and staff of the Family Cancer Clinics and the Clinical Follow Up Study (which has received funding from NHMRC, the National Breast Cancer Foundation, Cancer Australia and the National Institute of Health (USA)) for their contributions to this resource, and the many families who contribute to kConFab. We thank Peter MacCallum Cancer Centre Molecular Genomics core facility, supported by the Australian Cancer Research Foundation, and the Peter MacCallum Cancer Centre Bioinformatics core facility, in particular M. Jayawardana, for statistical advice. The WHIRC team acknowledge contributions by P. Akowuah, J. Loffredo, U. Rao, S. Makohon-Moore, J. Oliver, D. Mitchell and G. Gist. We thank the BC Cancer Foundation, Genome BC, Canadian Institutes for Health Research, Canadian Cancer Society, Terry Fox Research Institute and Canada Foundation for Innovation. Disclaimer: the views expressed herein are those of the authors and do not reflect the official policy of the Department of Army/Navy/Air Force, Department of Defense or US Government.

Author information

Authors and Affiliations

Authors

Contributions

E.L.C., K.A. and D.D.L.B. conceived the project and directed the study, with input from all authors. A.H., O.M., L.M., S.A., C.L.S., Y.A. and G.A.-Y. consented patients to CASCADE. N.L.B., M.O.W., K.A., K.P., J.H., L.D., H.T., D.D.L.B. and E.L.C. collected and/or processed patient tissues. N.L.B., K.A. and S.F. analyzed clinical information. For DNA- and RNA-seq, E.L.C., G.A.-Y. and D.D.L.B. contributed to the experimental design, M.O.W., S.C. and N.L.B. performed library preparation, A.P. performed data processing and N.L.B., M.O.W., T.H., S.C., K.I.P., D.W.G. and E.L.C. analyzed and interpreted data. X.L. and J.T.C. performed clonal analysis and interpretation. A.L.H., T.A., B.L.H., K.N.W., K.A.C., N.W.B., G.L.M. and T.P.C. optimized, performed and interpreted proteomics experiments and wrote the relevant section of the manuscript. P.T.H., K.M., S.K., A.M. and B.H.N. optimized, performed and interpreted mcIF experiments. A.B. and D.W.G. provided intellectual input and guidance on DNA- and RNA-seq analyses. N.L.B., E.L.C. and D.D.L.B. wrote the manuscript, with input from all authors.

Corresponding author

Correspondence to Elizabeth L. Christie.

Ethics declarations

Competing interests

T.P.C. is a ThermoFisher Scientific, Inc. SAB member and receives research funding from AbbVie. G.A.-Y. receives institutional grant funding from AstraZeneca and Roche-Genentech for unrelated work. AOCS (D.D.L.B., K.A., S.F. and J.H.) has received grant funding from AstraZeneca for an unrelated study. C.L.S. receives grant or research support from AstraZeneca, Clovis Oncology, Eisai, Inc., Sierra Oncology, Roche and Beigene for unrelated work, and sits on advisory boards for AstraZeneca, Clovis Oncology, Eisai, Inc., Sierra Oncology, Roche, Takeda and MSD. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Geoff Macintyre and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 CA125 and treatment data.

The clinical journey is shown for each patient, in 100 day intervals (x-axis), with the first tick referring to the day of first diagnostic information (surgery or CA125). Red line in the top plot indicates CA125 levels (y-axis), and triangles directly below indicate timing of primary surgery and death. The colored bars below indicate the treatments received and treatment duration.

Extended Data Fig. 2 Spatial sampling and distribution of mutations across samples.

(a) Top: simulated data demonstrating chance of detecting a variant present at frequencies between 0–1 (x axis) by the number of samples analyzed (y axis) (p < 0.0000000000000002, glm, t-value 10.62). Bottom: probability of accurate sampling based on the balance of tumor sampling (x axis) per number of samples (y axis) (p = <0.0000000000000002, glm, t-value 11.86). (b) Dendogram with oncoprint for autopsy only mutations within patients with a resistance mechanism detected in the targeted sequencing (that is, reversions and TP53BP1 INDEL), demonstrating their subclonal distribution. Each row represents a mutation within that patient, pink squares indicate mutation present, blue mutation absent. Only patients with a primary sample (to thereby distinguish mutations exclusive to autopsy setting) are shown.

Extended Data Fig. 3 Heterogeneity across WGS analyses.

(a) Copy number profiles for all samples ordered by chromosome (x-axis). Dendogram colored by first cluster split; pink encapsulates all non-whole genome duplicated samples. Copy number increases (red) and decreases (blue). (b) Example of the SV landscape as shown by circos plots for 6 autopsy samples from patient 15292. Lines indicate rearrangements and colors represent chromosomes. (c) Four cases with CCNE1 amplification in at least one tumor sample (all samples from those cases shown); the level of amplification is shown in each sample with the total copy number listed adjacently, with the total reversion allele frequency from the targeted sequencing (right). (d) Scatterplots of percent of autosomal genome duplicated vs TMB (top) and SV count (bottom). Samples in the red ellipse have WGD (>50% autosomal genome duplicated, n = 43), samples in the blue ellipse do not (n = 11).

Extended Data Fig. 4 Transcriptomic QC and analyses.

(a) Principal component analysis of RNAseq samples by QC parameters to assess for outlying samples with substandard parameters. Dimensions include = Dimension 1 < 2, Dimension 2 −0.4 > × < 1.8. Each panel shows the same PCA plot with samples colored by the following variables: RIN – RNA integrity number, TTA – time to autopsy, Uniquely mapped – number of uniquely mapped reads, Median CV coverage – variability of coverage over single genes, 3 prime bias (Picard tools), 5 prime bias (Picard tools). Color bar represents the numerical value of that graph title. (b) Sashimi plot schematic for case 66462 at BRCA1 exons 10–12 (Human genome reference build GRCh38 nomenclature used), showing the proportion of RNAseq reads comprising the canonical and delta 11q splice isoforms. Red arrow marks position of germline BRCA1 mutation. (c) MRE11 expression by sample MRE11 copy number state – RNAseq Z scores (p = 0.0002, glmm, z-statistic −3.75; amplified samples n = 3, non-amplified n = 17) and MRE11 protein abundance (p = 0.23, glm; t-value −1.25; amplified samples n = 5, non-amplified samples n = 97); blue represents copy neutral or loss, pink represents any level amplification. Lower and upper whisker terminates at the minimum and maximum values no further than 1.5 times the interquartile range; center line represents median (50th percentile); lower and upper boundary of box represent the 1st (25th percentile) and 3rd (75th percentile) quartiles respectively; outlying values are plotted as individual points beyond whiskers.

Source data

Extended Data Fig. 5 Evolution of immune composition in HGSC samples.

(a) McIF sample numbers by cores and metastatic sites per patient. (b) Stromal immune cell abundance (percentage of total cell detections) by timepoint. Left and right whisker terminates at the minimum and maximum values no further than 1.5 times interquartile range; center line represents median (50th percentile); left and right boundary of box represents the 1st (25th percentile) and 3rd (75th percentile) quartiles respectively. P values calculated with glmm, adjusted for multiple testing correction. Cut-off for significance accepted as p < 0.01; full statistics reported in Supplementary table 4. NA values occur where modelling could not be appropriately fitted due to small eigenvalues. X axis is logarithmic for improved visualization. Cores N = 282 primary, 488 autopsy (T&B cell) and 283 primary, 500 autopsy (PD1/MΦ). (c) Top: Boxplots of immune cell percentages within autopsy samples by patient. Bottom: variance of immune cell abundance between autopsy metastases per patient. For top and bottom, cores n = 488 (T&B cell) and 501 (PD1/MΦ). Lower and upper whisker terminates at minimum and maximum values no further than 1.5 times interquartile range; lower and upper boundary of box represent the 1st and 3rd quartiles; outlying values are plotted as individual points beyond whiskers; all others as in (b). (d) For 45 samples with both WGS and mcIF, bars represent the number of samples with or without loss of heterozygosity (LOH) of HLA alleles in WGS by mcIF cluster; samples which have cores in multiple mcIF clusters represented multiple times (p = 0.19, Chi sq, one-sided test).

Extended Data Fig. 6 Immune response and epigenetic regulation.

(a) Transcriptomic correlations between constitutive chemokines CCL5 and CXCL9 and T cell marker CD8A. N = 29 samples. Correlation statistics (R of repeated measures (rrm)) are labelled above plots. CCL5 vs CXCL9 Confidence interval (CI) = −0.03–0.78, 16 degrees of freedom; CCL5 vs CD8A CI = 0.20–0.86, 16 degrees of freedom; CXCL9 vs CD8A CI = 0.39–0.91, 16 degrees of freedom. (b) CCL5 expression (left; p = 0.0000000005; glmm, z-value = 6.2)), and M2 macrophage prevalence (right; p = 0.001; glmm, z-value = −3.191) by CCL5 methylation status (n = 21). Lower and upper whisker terminates at the minimum and maximum values no further than 1.5 times the interquartile range; center line represents median (50th percentile); lower and upper boundary of box represent the 1st (25th percentile) and 3rd (75th percentile) quartiles respectively; outlying values are plotted as individual points beyond whiskers. (c) PCA plot constructed from CCL5 and CXCL9 expression, mcIF CD8 T cell percentage and M2 macrophage abundance; left = annotated by RNA sample, right = annotated by anatomical site, demonstrating the overlap. RNAseq and methylation n = 19, corresponding to 42 mcIF cores. Dim = Principal component dimension.

Extended Data Fig. 7 Technical qualitative analyses of LMD enriched proteomics data.

(a) Workflow diagram depicting cell type enrichment via LMD and proteomic data analysis. Representative images from an H&E-stained tissue section and following LMD enrichment of tumor (blue) and stroma (orange) are shown (4 mm scale bar). (b) Supervised analysis of 107 differentially expressed proteins relating to the delay between patient death and specimen acquisition (p < 0.01, unadjusted for multiple testing correction, limma regression) between autopsy specimens (patient-blocked) relative to time to autopsy in LMD enriched tumor samples. Individual proteins are reported in Supplementary table 42. (c) Pearson correlation values of proteins related to previously defined signatures of necrosis and/or hypoxia in LMD enriched tumor samples. Shaded grey area represents 95% confidence interval.

Extended Data Fig. 8 Dendrograms demonstrating temporo-spatial proteomic differences.

Patient-specific phylogenetic analyses for 9 cases using Pearson correlations between early versus autopsy samples in LMD enriched tumor (left) or LMD enriched stroma (right). Timepoint abbreviations are primary debulking surgery (P), interval debulking surgery (I), recurrent sample (R), and autopsy sample (A). Patients with at least 2 early (non-autopsy) samples are included: (a) 15317, (b) 65659, (c) 15292, (d) 65682, (e) 15306, (f) 65913, (g) 66142, (h) 65938, and (i) 66462.

Supplementary information

Supplementary Information

Supplementary note, figures and associated references.

Reporting Summary

Supplementary Table 1

Supplementary tables for manuscript and text.

Source data

Source Data Fig. 5b

Clustering mcIF image (T and B cell panel, Imm-des) from composite image.

Source Data Fig. 5b

Clustering mcIF image (T and B cell panel, Ep/S infiltrated) from composite image

Source Data Fig. 5b

Clustering mcIF image (T and B cell panel, St-re) from composite image.

Source Data Fig. 5b

Clustering mcIF image (no. 65,914 PD1 panel) from composite image.

Source Data Fig. 5b

Clustering mcIF image (no. 65,914 PD1 panel) from composite image.

Source Data Extended Data Fig. 4b

IGV screenshot showing alternative splicing of BRCA1, from which schematic was created.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Burdett, N.L., Willis, M.O., Alsop, K. et al. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer. Nat Genet 55, 437–450 (2023). https://doi.org/10.1038/s41588-023-01320-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-023-01320-2

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research