The genomic landscape and evolution of endometrial carcinoma progression and abdominopelvic metastasis

William J Gibson, Erling A Hoivik, Mari K Halle, Amaro Taylor-Weiner, Andrew D Cherniack, Anna Berg, Frederik Holst, Travis I Zack, Henrica M J Werner, Kjersti M Staby, Mara Rosenberg, Ingunn M Stefansson, Kanthida Kusonmano, Aaron Chevalier, Karen K Mauland, Jone Trovik, Camilla Krakstad, Marios Giannakis, Eran Hodis, Kathrine Woie, Line Bjorge, Olav K Vintermyr, Jeremiah A Wala, Michael S Lawrence, Gad Getz, Scott L Carter, Rameen Beroukhim, Helga B Salvesen, William J Gibson, Erling A Hoivik, Mari K Halle, Amaro Taylor-Weiner, Andrew D Cherniack, Anna Berg, Frederik Holst, Travis I Zack, Henrica M J Werner, Kjersti M Staby, Mara Rosenberg, Ingunn M Stefansson, Kanthida Kusonmano, Aaron Chevalier, Karen K Mauland, Jone Trovik, Camilla Krakstad, Marios Giannakis, Eran Hodis, Kathrine Woie, Line Bjorge, Olav K Vintermyr, Jeremiah A Wala, Michael S Lawrence, Gad Getz, Scott L Carter, Rameen Beroukhim, Helga B Salvesen

Abstract

Recent studies have detailed the genomic landscape of primary endometrial cancers, but the evolution of these cancers into metastases has not been characterized. We performed whole-exome sequencing of 98 tumor biopsies including complex atypical hyperplasias, primary tumors and paired abdominopelvic metastases to survey the evolutionary landscape of endometrial cancer. We expanded and reanalyzed The Cancer Genome Atlas (TCGA) data, identifying new recurrent alterations in primary tumors, including mutations in the estrogen receptor cofactor gene NRIP1 in 12% of patients. We found that likely driver events were present in both primary and metastatic tissue samples, with notable exceptions such as ARID1A mutations. Phylogenetic analyses indicated that the sampled metastases typically arose from a common ancestral subclone that was not detected in the primary tumor biopsy. These data demonstrate extensive genetic heterogeneity in endometrial cancers and relative homogeneity across metastatic sites.

Conflict of interest statement

Competing Financial Interests

The authors declare no competing financial interests.

Figures

Figure 1. Samples assessed
Figure 1. Samples assessed
(a) Anatomic sites from which samples were obtained. (b) Histologic subtypes (EN: endometrioid, CC: clear cell, S: serous and U: undifferentiated carcinomas, and CS: carcinosarcoma), grade, FIGO 2009 stage at primary diagnosis, time of metastatic lesion sampling, and treatment after primary diagnosis. Asterisks indicate four cases that were clinically difficult to distinguish as metastatic or independent synchronous primary cancers at time of resection.
Figure 2. Somatic genetic alterations in complex…
Figure 2. Somatic genetic alterations in complex atypical hyperplasias and primary and metastatic endometrial carcinomas
(a) Number of exonic mutations (top) and SCNAs (middle) detected in each tumor biopsy. PTEN, TP53, and 1q amplification status, histologic subtype, TCGA annotation, and tissue type are indicated on the bottom. (b) Number of mutations (y-axis) against fraction of the genome affected by SCNAs (x-axis) across complex atypical hyperplasias (green), primary lesions from endometrioid endometrial carcinoma (red) and non-endometrioid endometrial carcinoma (blue); primaries from our dataset (squares) and TCGA (dots). (c) Number of mutations detected in the primary tumor compared to their metastatic counterpart. (d) Fraction of the genome affected by SCNAs in metastases (y-axis) relative to paired primaries (x-axis). Circles indicate metastases that exhibit whole-genome doubling not observed in the primary biopsy. (e) Stick plot depicting detected mutations in NRIP1, a cofactor of the estrogen receptor. (f) Impact of indel rescue on the percentage of patients harboring mutations in known driver genes.
Figure 3. Heterogeneity among somatic mutations
Figure 3. Heterogeneity among somatic mutations
Fractions of (a) all mutations and (b) driver mutations detected in metastases that were truncal. Lines indicate the mean. (c) The number truncal and branch mutations involving the indicated driver genes. (d) Percentage of mutations that were detected in all paired biopsies in our dataset for frequently mutated driver genes. ARID1A is frequently mutated in the branches of phylogenies. (e) Distribution of cancer cell fraction (CCF) values for each detected mutation in TCGA endometrial samples for the indicated genes. Bars indicate 95% confidence intervals. (f) Distribution of the probability that each mutation detected in TCGA endometrial biopsies is clonal for the indicated genes. Hypermutated samples (> 1,000 mutations) were excluded from (e) and (f)
Figure 4. Phylogenetic trees for tumors with…
Figure 4. Phylogenetic trees for tumors with more than one metastasis
(a) The labeled alterations constitute a subset of the alterations that distinguish between the indicated branches. Hash symbols indicate trees that were derived from SNP6.0 array data. (b) 2D phylogenetic illustration comparing subclonal structures between biopsies in patient EC-007. Cancer cell fraction (CCF) probability densities for the mutations supporting the metastasizing subclone (purple) are presented in Supplementary Figure 16. (c) Phylogenetic tree of patient EC-007. A subclone in the biopsy of the primary tumor shared a common ancestor with the two metastases, not detected in the dominant clone of the primary biopsy. Subclone 2 from (b) could be placed in three different phylogenetic locations as indicated by the dotted purple lines.

References

    1. Bhaskaran K, et al. Body-mass index and risk of 22 specific cancers: a population-based cohort study of 5.24 million UK adults. Lancet. 2014;384:755–765.
    1. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. 1983;15:10–17.
    1. Cancer Genome Atlas Research, N. et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497:67–73.
    1. Salvesen HB, et al. Integrated genomic profiling of endometrial carcinoma associates aggressive tumors with indicators of PI3 kinase activation. Proc Natl Acad Sci U S A. 2009;106:4834–4839.
    1. Dutt A, et al. Drug-sensitive FGFR2 mutations in endometrial carcinoma. Proc Natl Acad Sci U S A. 2008;105:8713–8717.
    1. Salvesen HB, Haldorsen IS, Trovik J. Markers for individualised therapy in endometrial carcinoma. Lancet Oncol. 2012;13:e353–e361.
    1. Ciriello G, et al. Emerging landscape of oncogenic signatures across human cancers. Nat Genet. 2013;45:1127–1133.
    1. Giannakis M, et al. RNF43 is frequently mutated in colorectal and endometrial cancers. Nat Genet. 2014;46:1264–1266.
    1. Lawrence MS, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495–501.
    1. Woodruff JD, Pickar JH. Incidence of endometrial hyperplasia in postmenopausal women taking conjugated estrogens (Premarin) with medroxyprogesterone acetate or conjugated estrogens alone. The Menopause Study Group. Am J Obstet Gynecol. 1994;170:1213–1223.
    1. Veeraraghavan J, et al. Recurrent ESR1-CCDC170 rearrangements in an aggressive subset of oestrogen receptor-positive breast cancers. Nat Commun. 2014;5:4577.
    1. Cavailles V, et al. Nuclear factor RIP140 modulates transcriptional activation by the estrogen receptor. EMBO J. 1995;14:3741–3751.
    1. Rosell M, et al. Complex formation and function of estrogen receptor alpha in transcription requires RIP140. Cancer Res. 2014;74:5469–5479.
    1. Toy W, et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet. 2013;45:1439–1445.
    1. Robinson DR, et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet. 2013;45:1446–1451.
    1. Kamburov A, et al. Comprehensive assessment of cancer missense mutation clustering in protein structures. Proc Natl Acad Sci U S A. 2015;112:E5486–E5495.
    1. Chang MT, et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat Biotechnol. 2015
    1. Novetsky AP, et al. Frequent mutations in the RPL22 gene and its clinical and functional implications. Gynecol Oncol. 2013;128:470–474.
    1. Hong B, Le Gallo M, Bell DW. The mutational landscape of endometrial cancer. Curr Opin Genet Dev. 2015;30:25–31.
    1. Ren Y, et al. JAK1 truncating mutations in gynecologic cancer define new role of cancer-associated protein tyrosine kinase aberrations. Sci Rep. 2013;3:3042.
    1. Hayes MP, et al. PIK3CA and PTEN mutations in uterine endometrioid carcinoma and complex atypical hyperplasia. Clin Cancer Res. 2006;12:5932–5935.
    1. Brastianos P, et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discovery. 2015
    1. Carter SL, et al. Absolute quantification of somatic DNA alterations in human cancer. Nature biotechnology. 2012;30:413–421.
    1. Ganem NJ, Godinho SA, Pellman D. A mechanism linking extra centrosomes to chromosomal instability. Nature. 2009;460:278–282.
    1. Zack TI, et al. Pan-cancer patterns of somatic copy number alteration. Nat Genet. 2013;45:1134–1140.
    1. Stachler M, et al. Paired Exome Analysis of Barrett’s Esophagus and Adenocarcinoma. Nature Genetics. 2015
    1. Gundem G, et al. The evolutionary history of lethal metastatic prostate cancer. Nature. 2015
    1. Wright JD, Barrena Medel NI, Sehouli J, Fujiwara K, Herzog TJ. Contemporary management of endometrial cancer. Lancet. 2012;379:1352–1360.
    1. Mao TL, et al. Loss of ARID1A expression correlates with stages of tumor progression in uterine endometrioid carcinoma. Am J Surg Pathol. 2013;37:1342–1348.
    1. Landau, Dan A, et al. Evolution and Impact of Subclonal Mutations in Chronic Lymphocytic Leukemia. Cell. 2013;152:714–726.
    1. Lohr JG, et al. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy. Cancer Cell. 2014;25:91–101.
    1. Ding L, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075.
    1. Zhao S, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci U S A. 2013;110:2916–2921.
    1. Campbell PJ, et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467:1109–1113.
    1. Yachida S, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–1117.
    1. McFadden DG, et al. Genetic and clonal dissection of murine small cell lung carcinoma progression by genome sequencing. Cell. 2014;156:1298–1311.
    1. Ghoussaini M, et al. Genome-wide association analysis identifies three new breast cancer susceptibility loci. Nat Genet. 2012;44:312–318.
    1. Gerlinger M, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46:225–233.
    1. Johnson BE, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science. 2014;343:189–193.
    1. Abedalthagafi MS, et al. ARID1A and TERT promoter mutations in dedifferentiated meningioma. Cancer Genet. 2015
    1. Torres-Martin M, et al. Whole exome sequencing in a case of sporadic multiple meningioma reveals shared NF2, FAM109B, and TPRXL mutations, together with unique SMARCB1 alterations in a subset of tumor nodules. Cancer Genet. 2015
    1. Lee RS, et al. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J Clin Invest. 2012;122:2983–2988.
    1. Bitler BG, et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med. 2015;21:231–238.
    1. Werner HM, et al. ARID1A loss is prevalent in endometrial hyperplasia with atypia and low-grade endometrioid carcinomas. Mod Pathol. 2013;26:428–434.
    1. Wiegand KC, et al. Loss of BAF250a (ARID1A) is frequent in high-grade endometrial carcinomas. J Pathol. 2011;224:328–333.
    1. Kim MY, et al. Tumor self-seeding by circulating cancer cells. Cell. 2009;139:1315–1326.
    1. Valastyan S, Weinberg RA. Tumor metastasis: molecular insights and evolving paradigms. Cell. 2011;147:275–292.
Methods-only References
    1. Berg A, et al. Molecular profiling of endometrial carcinoma precursor, primary and metastatic lesions suggests different targets for treatment in obese compared to non-obese patients. Oncotarget. 2015;6:1327–1339.
    1. Wik E, et al. Endometrial Carcinoma Recurrence Score (ECARS) validates to identify aggressive disease and associates with markers of epithelial-mesenchymal transition and PI3K alterations. Gynecol Oncol. 2014;134:599–606.
    1. Saunders CT, et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics. 2012;28:1811–1817.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nature biotechnology. 2013;31:213–219.
    1. Costello M, et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Res. 2013;41:e67.
    1. Zhu S, Degnan JH, Steel M. Clades, clans, and reciprocal monophyly under neutral evolutionary models. Theor Popul Biol. 2011;79:220–227.

Source: PubMed

3
Abonnieren