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.

Whole–genome characterization of chemoresistant ovarian cancer

A Corrigendum to this article was published on 21 October 2015

Abstract

Patients with high-grade serous ovarian cancer (HGSC) have experienced little improvement in overall survival, and standard treatment has not advanced beyond platinum-based combination chemotherapy, during the past 30 years. To understand the drivers of clinical phenotypes better, here we use whole-genome sequencing of tumour and germline DNA samples from 92 patients with primary refractory, resistant, sensitive and matched acquired resistant disease. We show that gene breakage commonly inactivates the tumour suppressors RB1, NF1, RAD51B and PTEN in HGSC, and contributes to acquired chemotherapy resistance. CCNE1 amplification was common in primary resistant and refractory disease. We observed several molecular events associated with acquired resistance, including multiple independent reversions of germline BRCA1 or BRCA2 mutations in individual patients, loss of BRCA1 promoter methylation, an alteration in molecular subtype, and recurrent promoter fusion associated with overexpression of the drug efflux pump MDR1.

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: Genomic features of HGSC.
Figure 2: Mutational signatures of primary high-grade serous ovarian cancer.
Figure 3: Somatic mutational patterns in acquired resistant cohort.
Figure 4: Molecular changes associated with acquired resistance.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

Data deposits

The whole genome and transcriptome sequencing data have been deposited in the European Genome-phenome Archive (EGA) repository under the accession code EGAD00001000877. Genotyping, methylation and miRNA data sets have been submitted into the Gene Expression Omnibus (GEO) accession GSE65821. A complete list of the AOCS Study Group can be found at http://www.aocstudy.org.

References

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

    Article  CAS  Google Scholar 

  2. 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 

  3. The Cancer Genome Atlas Research Network. Integrated genomic analysis of ovarian cancer. Nature 474, 609–615 (2011).

  4. Gorringe, K. L. et al. High-resolution single nucleotide polymorphism array analysis of epithelial ovarian cancer reveals numerous microdeletions and amplifications. Clin. Cancer Res. 13, 4731–4739 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Ciriello, G. et al. Emerging landscape of oncogenic signatures across human cancers. Nature Genet. 45, 1127–1133 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Mukhopadhyay, A. et al. Development of a functional assay for homologous recombination status in primary cultures of epithelial ovarian tumor and correlation with sensitivity to poly(ADP-ribose) polymerase inhibitors. Clin. Cancer Res. 16, 2344–2351 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. Castellarin, M. et al. Clonal evolution of high-grade serous ovarian carcinoma from primary to recurrent disease. J. Pathol. 229, 515–524 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Cowin, P. A. et al. LRP1B deletion in high-grade serous ovarian cancers is associated with acquired chemotherapy resistance to liposomal doxorubicin. Cancer Res. 72, 4060–4073 (2012).

    Article  CAS  PubMed  Google Scholar 

  10. Popova, T. et al. Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation. Cancer Res. 72, 5454–5462 (2012).

    Article  CAS  PubMed  Google Scholar 

  11. George, J. et al. Nonequivalent gene expression and copy number alterations in high-grade serous ovarian cancers with BRCA1 and BRCA2 mutations. Clin. Cancer Res. 19, 3474–3484 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Clarke, B. et al. Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod. Pathol. 22, 393–402 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Soslow, R. A. et al. Morphologic patterns associated with BRCA1 and BRCA2 genotype in ovarian carcinoma. Mod. Pathol. 25, 625–636 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kinsella, M. & Bafna, V. Combinatorics of the breakage-fusion-bridge mechanism. J. Comput. Biol. 19, 662–678 (2012).

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  16. Baca, S. C. et al. Punctuated evolution of prostate cancer genomes. Cell 153, 666–677 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rausch, T. et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 148, 59–71 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Nones, K. et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int. J. Cancer 135, 1110–1118 (2014).

    Article  CAS  PubMed  Google Scholar 

  19. Salzman, J. et al. ESRRA-C11orf20 is a recurrent gene fusion in serous ovarian carcinoma. PLoS Biol. 9, e1001156 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kannan, K. et al. CDKN2D-WDFY2 is a cancer-specific fusion gene recurrent in high-grade serous ovarian carcinoma. PLoS Genet. 10, e1004216 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Etemadmoghadam, D. et al. Integrated genome-wide DNA copy number and expression analysis identifies distinct mechanisms of primary chemoresistance in ovarian carcinomas. Clin. Cancer Res. 15, 1417–1427 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Verhaak, R. G. et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J. Clin. Invest. 123, 517–525 (2013).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Lengyel, E. Ovarian cancer development and metastasis. Am. J. Pathol. 177, 1053–1064 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Swisher, E. M. et al. Secondary BRCA1 mutations in BRCA1-mutated ovarian carcinomas with platinum resistance. Cancer Res. 68, 2581–2586 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Olive, K. P. et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 324, 1457–1461 (2009).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Geisler, J. P., Hatterman-Zogg, M. A., Rathe, J. A. & Buller, R. E. Frequency of BRCA1 dysfunction in ovarian cancer. J. Natl. Cancer Inst. 94, 61–67 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Meier, B. et al. C. elegans whole-genome sequencing reveals mutational signatures related to carcinogens and DNA repair deficiency. Genome Res. 24, 1624–1636 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Edwards, S. L. et al. Resistance to therapy caused by intragenic deletion in BRCA2. Nature 451, 1111–1115 (2008).

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Sakai, W. et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature 451, 1116–1120 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Vergote, I. et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N. Engl. J. Med. 363, 943–953 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Murray, S., Briasoulis, E., Linardou, H., Bafaloukos, D. & Papadimitriou, C. Taxane resistance in breast cancer: mechanisms, predictive biomarkers and circumvention strategies. Cancer Treat. Rev. 38, 890–903 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Binkhathlan, Z. & Lavasanifar, A. P-glycoprotein inhibition as a therapeutic approach for overcoming multidrug resistance in cancer: current status and future perspectives. Curr. Cancer Drug Targets 13, 326–346 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Lhommé, C. et al. Phase III study of valspodar (PSC 833) combined with paclitaxel and carboplatin compared with paclitaxel and carboplatin alone in patients with stage IV or suboptimally debulked stage III epithelial ovarian cancer or primary peritoneal cancer. J. Clin. Oncol. 26, 2674–2682 (2008).

    Article  PubMed  CAS  Google Scholar 

  38. Jaspers, J. E. et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov. 3, 68–81 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Johnson, B. E. et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343, 189–193 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  40. Lawrence, M. S. et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 505, 495–501 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Rustin, G. J. et al. Definitions for response and progression in ovarian cancer clinical trials incorporating RECIST 1.1 and CA 125 agreed by the Gynecological Cancer Intergroup (GCIG). Int. J. Gynecol. Cancer 21, 419–423 (2011).

    Article  PubMed  Google Scholar 

  42. Friedlander, M. L. et al. Clinical trials of palliative chemotherapy in platinum-resistant or -refractory ovarian cancer: time to think differently? J. Clin. Oncol. 31, 2362 (2013).

    Article  PubMed  Google Scholar 

  43. Popova, T. et al. Genome Alteration Print (GAP): a tool to visualize and mine complex cancer genomic profiles obtained by SNP arrays. Genome Biol. 10, R128 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Mermel, C. H. et al. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 12, R41 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  48. DeLuca, D. S. et al. RNA-SeQC: RNA-seq metrics for quality control and process optimization. Bioinformatics 28, 1530–1532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, K. et al. MapSplice: accurate mapping of RNA-seq reads for splice junction discovery. Nucleic Acids Res. 38, e178 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Wilhelm-Benartzi, C. S. et al. Review of processing and analysis methods for DNA methylation array data. Br. J. Cancer 109, 1394–1402 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  PubMed  MATH  Google Scholar 

  53. Kassahn, K. S. et al. Somatic point mutation calling in low cellularity tumors. PLoS ONE 8, e74380 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  54. McKenna, A. et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Lawrence, M. S. et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499, 214–218 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Gundem, G. et al. IntOGen: integration and data mining of multidimensional oncogenomic data. Nature Methods 7, 92–93 (2010).

    Article  CAS  PubMed  Google Scholar 

  57. Ye, K., Schulz, M. H., Long, Q., Apweiler, R. & Ning, Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics 25, 2865–2871 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Robinson, J. T. et al. Integrative genomics viewer. Nature Biotechnol. 29, 24–26 (2011).

    Article  CAS  Google Scholar 

  59. Ojesina, A. I. et al. Landscape of genomic alterations in cervical carcinomas. Nature 506, 371–375 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  60. 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 

  61. Korbel, J. O. & Campbell, P. J. Criteria for inference of chromothripsis in cancer genomes. Cell 152, 1226–1236 (2013).

    Article  CAS  PubMed  Google Scholar 

  62. Koboldt, D. C. et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The AOCS gratefully acknowledge the cooperation of the participating institutions in Australia, and also acknowledge the contribution of the study nurses, research assistants and all clinical and scientific collaborators including L. Galletta, C. Emmanuel, L. Bowes and J. Hallo. The authors acknowledge assistance from C. Anderson and D. Gwynne. The CASCADE investigators would like to thank the CASCADE Management Committee, all staff at the Victorian Institute of Forensic Medicine, D. Stevens and Tobin Brothers Funerals. The investigators would like to thank the Australia New Zealand Gynaecological Oncology Group (ANZGOG) and the women, and their families, who participated in these research programs. This work was supported by the National Health and Medical Research Council of Australia (NHMRC ID631701), Worldwide Cancer Research (09-0676) and Cancer Australia (1004673). The Australian Ovarian Cancer Study was supported by the US Army Medical Research and Materiel Command under DAMD17-01-1-0729, The Cancer Council Victoria, Queensland Cancer Fund, The Cancer Council New South Wales, The Cancer Council South Australia, The Cancer Foundation of Western Australia, The Cancer Council Tasmania and the National Health and Medical Research Council of Australia (NHMRC; ID400413, ID400281). The AOCS gratefully acknowledges additional support from S. Boldeman, the Agar family, Ovarian Cancer Australia and Ovarian Cancer Action (UK). The Gynaecological Oncology Biobank at Westmead, a member of the Australasian Biospecimen Network-Oncology group, was supported by grants from the NHMRC (ID 310670, ID 628903) and the Cancer Institute of New South Wales. 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.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Project supervision: J.V.P., Nicola.W., A.D.F., S.M.G., D.D.L.B. Study design: A.M.P., E.L.C., D.E., D.W.G., S.F., P.C., Nicola.W., A.D.F., S.M.G., D.D.L.B. Sample acquisition: S.F., K.A., A.O.C.S., A.H., L.M., O.M.N., C.K., J.H., Y.E.C., P.H., M.F., M.Q., J.P., S.C., P.O.B., J.L., P.W., N.T., H.T., M.S., R.D., E.S., H.G., A.J., O.M.N., E.L., A.D.F., D.D.L.B. Sample preparation: E.L.C., D.E., D.W.G., P.C., Y.E.C., P.H., C.M., J.H. Data acquisition: E.L.C., D.E., D.W.G., S.F., D.K.M., G.M.A., T.P.H., T.S., I.H., C.N., E.N., S.M., S.I., T.J.C.B., A.N.C. Performed patient autopsy: G.Y., K.S. Sequence data management, alignment and mutation identification: A.M.P., K.N., F.N., S.K., O.H., M.A., C.L., S.W., Q.X., J.L.F., Nick.W., S.H.N., P.J.W., J.V.P., S.M.G. Genome informatics, software tool development: A.M.P., P.J.B., K.S.K., F.N., S.K., B.P., O.H., M.A., C.L., S.W., D.F.T., Q.X., J.L.F., Nick.W., J.V.P., Nicola.W. Data analysis: A.M.P., E.L.C., D.E., D.W.G., J.G., K.N., P.J.B., K.S.K., M.C.JQ., K.Q., C.W.B., E.C., H.S.L., G.A.Y., W.A., R.W.T., A.L., N.H., R.B., J.E., M.D., R.V., C.S., J.V.P., Nicola.W., S.M.G. Perform molecular/verification analysis: E.L.C., K.N., S.M., E.M. Wrote the manuscript: A.M.P., E.L.C., D.E., D.W.G., Nicola.W., A.D.F., S.M.G., D.D.L.B.

Corresponding author

Correspondence to David D. L. Bowtell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

A list of authors and affiliations appears in the Supplementary Information

Extended data figures and tables

Extended Data Figure 1 Patient cohort.

a, Summary of whole-genome sequenced patients (n = 92) and samples (n = 114). b, Clinical characteristics of patients by clinical response group and acquired resistant cases without matching primary tumour material. *Primary tumour ungraded; diagnosis from ascites or pleural fluid. ^Time to progression or death measured from diagnosis. aKruskal–Wallis, bFisher or clog-rank test P values comparing primary tumour clinical groups reported. Median follow up time of cohort was 97.3 months.

Extended Data Figure 2 Structural variants and somatic mutations.

a, b, Scatter plot shows that the number of structural variants (a) and SNVs (b) detected per sample was not dependent on the purity of primary tumours (n = 80, grey) and ascites/relapse samples (n = 34, black). Spearman correlation P values indicated. c, Number of structural variants detected in primary tumours (n = 80) grouped by homologous recombination (HR) mutation status: HR wild-type, BRCA1/2 altered, and HR deficient (lines indicate mean; ****P < 0.0001, Kolmogorov–Smirnov test). d, e, Whole-genome (d) and coding mutations (e) per megabase for samples (n = 79) stratified by BRCA1, BRCA2 or homologous recombination pathway mutation status (lines indicate mean; **P < 0.01, ***P < 0.001, Kolmogorov–Smirnov test). Sample AOCS-166 with germline mismatch repair mutation excluded from analysis (see Supplementary Information section 4.3).

Extended Data Figure 3 Chromothripsis and breakage-fusion-bridge.

a, Chromothripsis affecting chromosome 13, including the BRCA2 locus (arrow), for a primary chemotherapy-resistant tumour. A high number of breakpoints (structural variants) are observed with oscillations of copy number (CNA and LOGRR) indicating regions of retained heterozygosity (BAF) for a single haplotype. b, Breakage-fusion-bridge amplification is observed on chromosome 1 for a primary chemotherapy-resistant tumour. A cluster of breakpoints (structural variants), mostly inversions, on the distal p arm are associated with blocks of amplification (CNA). IGV review of the tumour WGS confirms that the telomere region has been lost.

Extended Data Figure 4 Expression of genes altered by structural variation.

Scatter graphs show expression of RB1, NF1, PTEN and RAD51B plotted against copy number in primary tumours (n = 79; Spearman correlation analysis). Boxplots summarize expression by mutation type; lines indicate median and whiskers show range (two-tailed Mann-Whitney test; *P < 0.05, ***P < 0.001). Samples with somatic interrupting structural variants and truncating mutations are indicated. Not all structural variants are associated with DNA loss and translocations can be observed to produce high expression counts for non-functional transcripts. Expression values lower than the median value for tumours and controls was observed for the majority of samples with structural variant events in RB1, NF1, PTEN and RAD51B.

Extended Data Figure 5 Molecular characteristics of primary tumours.

Clinical and molecular characteristics of primary tumours sorted by BRCA signature contribution (n = 80).

Extended Data Figure 6 Mutational signature associations.

a, b, Age mutational signature association with age at diagnosis (a) and CCNE1 copy number (b) (Spearman correlation P value reported, n = 80). c, d, BRCA mutational signature association with BRCA1/2 and other homologous recombination pathway mutations (c) and primary treatment response (d) (lines indicate mean; Kolmogorov–Smirnov **P < 0.01, ***P < 0.001, ****P < 0.0001; n = 80).

Extended Data Figure 7 Molecular drivers and clinical associations.

a, Percentage (n) of primary tumours (total n = 80) affected by homologous recombination pathway mutations and CCNE1 copy number gains. One driver mutation counted for samples with more than one change, ranking mutations in BRCA1/2, followed by other germline, somatic, amplification, deletion and methylation events respectively. b, Association of driver mutation subgroup with overall survival in AOCS and TCGA cohorts (Kaplan–Meier analysis, P value calculated by Mantel–Cox log-rank test). c, d, Whole genome (c) and coding mutations (d) per megabase for samples stratified by primary clinical response group (lines indicate mean). Kruskal–Wallis test P value reported (*P < 0.05, **P < 0.01, Kolmogorov–Smirnov test). e, Boxplots summarize CCNE1 expression in different driver mutation subgroups (****P < 0.0001, unpaired two-tailed t-test). Middle bar, median; whiskers, data range. f, Proportions of gene expression molecular subtypes between driver mutation subgroups. HR/CCNE1− subgroup has no detected homologous recombination pathway mutations or CCNE1 copy number changes.

Extended Data Figure 8 Analysis of acquired resistance cases.

a, b, Matched primary ascites share most variants with primary tumour samples across the whole genome (a) and for non-silent coding mutations (b). c, d, Significant correlations are observed between the time between collection of the primary and relapse samples and the number of lines of platinum treatment the patient received (Pearson correlation, P = 0.0232, two-tailed) (c), and between the number of non-silent coding mutations unique to the relapse samples and the number of lines of platinum treatment the patient received (Pearson correlation, P = 0.0456, two-tailed) (d). e, Genes with recurrent non-silent coding mutations that are unique to the second sample in the acquired resistance cohort or that are driver genes reported previously55.

Extended Data Figure 9 BRCA1/2 reversion events.

a, BRCA1 and BRCA2 mutations are indicated by sample for acquired resistance cases (n = 23). Of the ten cases with germline mutations, five show secondary somatic mutations in the relapse-resistant samples that are proposed to restore gene function (that is, reversion). b, Two independent point mutations revert the BRCA1 nonsense mutation in AOCS-034. c, CA125 serum marker profile for case AOCS-167. d, Schematic of 12 high confidence reversion events identified in AOCS-167. e, Allele frequency of fifteen BRCA2 reversion events identified in deep amplicon sequencing across 18 relapse samples from case AOCS-167. Reversions observed in single deposits at low allele frequencies (≤0.001) were considered low confidence events.

Extended Data Figure 10 SLC25A40-ABCB1 fusion transcript.

a, Schematic showing the locations of structural variants identified upstream or internal to ABCB1 from WGS. b, ABCB1 and SLC25A40 expression levels in sensitive and resistant samples of AOCS-092 and AOCS-150. c, RT–PCR verification of SLC25A40-ABCB1 fusion transcript expression in AOCS-092 and schematic of expected RT–PCR products. d, RT–PCR results from a validation cohort of relapse ascites identifying four additional samples with the fusion transcript (n = 51). e, ABCB1 and SLC25A40 expression levels compared to the SKOV3 cell line by qRT–PCR in the validation cohort and selected cases. Relapse samples with the fusion are indicated by open squares and the matched primary samples indicated by closed squares; validation cases with the fusion are indicated by ‘+’; the median plus median absolute deviation expression level is indicated by the dotted line (n = 56).

Supplementary information

Supplementary Information

This file contains Supplementary Text, Data and Figures (see Contents list for details), a list of the AOCS Study Group Authors and Affiliations and additional references. (PDF 5691 kb)

Supplementary Tables

This zipped file contains Supplementary Tables 1-18. (ZIP 30672 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Patch, AM., Christie, E., Etemadmoghadam, D. et al. Whole–genome characterization of chemoresistant ovarian cancer. Nature 521, 489–494 (2015). https://doi.org/10.1038/nature14410

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14410

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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