The genetic landscape of endometrial clear cell carcinomas
Corresponding Author
Deborah F DeLair
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Correspondence to: DF DeLair, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or RA Soslow, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or B Weigelt, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected]Search for more papers by this authorKathleen A Burke
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorPier Selenica
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorRaymond S Lim
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorSasinya N Scott
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorSumit Middha
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorAbhinita S Mohanty
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorDonavan T Cheng
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorMichael F Berger
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorCorresponding Author
Robert A Soslow
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Correspondence to: DF DeLair, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or RA Soslow, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or B Weigelt, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected]Search for more papers by this authorCorresponding Author
Britta Weigelt
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Correspondence to: DF DeLair, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or RA Soslow, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or B Weigelt, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected]Search for more papers by this authorCorresponding Author
Deborah F DeLair
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Correspondence to: DF DeLair, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or RA Soslow, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or B Weigelt, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected]Search for more papers by this authorKathleen A Burke
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorPier Selenica
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorRaymond S Lim
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorSasinya N Scott
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorSumit Middha
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorAbhinita S Mohanty
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorDonavan T Cheng
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorMichael F Berger
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Search for more papers by this authorCorresponding Author
Robert A Soslow
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Correspondence to: DF DeLair, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or RA Soslow, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or B Weigelt, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected]Search for more papers by this authorCorresponding Author
Britta Weigelt
Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Correspondence to: DF DeLair, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or RA Soslow, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected] Or B Weigelt, Department of Pathology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA. E-mail: [email protected]Search for more papers by this authorAbstract
Clear cell carcinoma of the endometrium is a rare type of endometrial cancer that is generally associated with an aggressive clinical behaviour. Here, we sought to define the repertoire of somatic genetic alterations in endometrial clear cell carcinomas (ECCs), and whether ECCs could be classified into the molecular subtypes described for endometrial endometrioid and serous carcinomas. We performed a rigorous histopathological review, immunohistochemical analysis and massively parallel sequencing targeting 300 cancer-related genes of 32 pure ECCs. Eleven (34%), seven (22%) and six (19%) ECCs showed abnormal expression patterns for p53, ARID1A, and at least one DNA mismatch repair (MMR) protein, respectively. Targeted sequencing data were obtained from 30 of the 32 ECCs included in this study, and these revealed that two ECCs (7%) were ultramutated and harboured mutations affecting the exonuclease domain of POLE. In POLE wild-type ECCs, TP53 (46%), PIK3CA (36%), PPP2R1A (36%), FBXW7 (25%), ARID1A (21%), PIK3R1 (18%) and SPOP (18%) were the genes most commonly affected by mutations; 18% and 11% harboured CCNE1 and ERBB2 amplifications, respectively, and 11% showed DAXX homozygous deletions. ECCs less frequently harboured mutations affecting CTNNB1 and PTEN but more frequently harboured PPP2R1A and TP53 mutations than non-POLE endometrioid carcinomas from The Cancer Genome Atlas (TCGA). Compared to endometrial serous carcinomas (TCGA), ECCs less frequently harboured TP53 mutations. When a surrogate model for the molecular-based TCGA classification was used, all molecular subtypes previously identified in endometrial endometrioid and serous carcinomas were present in the ECCs studied, including POLE, MMR-deficient, copy-number high (serous-like)/p53 abnormal, and copy-number low (endometrioid)/p53 wild-type, which were significantly associated with disease-free survival in univariate analysis. These findings demonstrate that ECCs constitute a histologically and genetically heterogeneous group of tumours with varying outcomes. Furthermore, our data suggest that the classification of ECCs as being generally ‘high-grade’ or ‘type II’ tumours may not be warranted. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Supporting Information
Filename | Description |
---|---|
path4947-sup-0001-SuppMat.docxWord 2007 document , 149.4 KB | Supplementary materials and methods |
path4947-sup-0002-FigureS1.tifTIFF image, 686.4 KB | Figure S1. Likely pathogenic non-synonymous somatic mutations detected by targeted capture massively parallel sequencing in endometrial clear cell carcinomas. Recurrent (n≥2) likely pathogenic non-synonymous somatic mutations (top) and recurrent (n≥2) amplifications and homozygous deletions (bottom) identified in 28 POLE-wild-type endometrial clear cell carcinomas by massively parallel sequencing targeting 300 cancer-related genes. Cases are represented in columns; genes are depicted in rows. Mutation types and gene copy number alterations are color-coded according to the legend. Loss of heterozygosity of the wild-type allele in association with a somatic mutation is depicted by a diagonal bar. Two additional cases, CC26 and CC31, were found to harbor somatic POLE exonuclease domain mutations (EDM), and are shown on the right. The phenobar provides information on stage, POLE status, ARID1A and p53 immunohistochemistry, and is color-coded according to the legend. Indel, small insertion/ deletion; SNV, single nucleotide variant. |
path4947-sup-0003-FigureS2.tifTIFF image, 959.3 KB | Figure S2. Cancer cell fractions of non-synonymous somatic mutations detected by targeted capture massively parallel sequencing in endometrial clear cell carcinomas. Cancer cell fractions as defined by ABSOLUTE of recurrent (n≥2) non-synonymous somatic mutations identified in 28 POLE-wild-type endometrial clear cell carcinomas by massively parallel sequencing targeting 300 cancer-related genes are shown. Cases are represented in columns; genes are depicted in rows. Loss of heterozygosity of the wild-type allele in association with a somatic mutation is depicted by a diagonal bar, clonal mutations by an orange box. The phenobar provides information on stage, ARID1A and p53 immunohistochemistry, and is color-coded according to the legend. |
path4947-sup-0004-FigureS3.tifTIFF image, 4.6 MB | Figure S3. Histological features and copy number profiles of molecular subtypes of endometrial clear cell carcinomas. Using a surrogate model for the molecular-based TOGA classification for endometrial endometrioid and serous carcinomas [38], endometrial clear cell carcinomas (ECCs) were classified into POLE, MMR-deficient (MMR-D), p53 wild-type (copy-number low (endometrioid)) and p53 abnormal (copy-number high-like (serous-like)) sub-types. Representative hematoxylin and eosin stained sections are shown on the left and chromosome plots on the right, where Loge ratios were plotted on the y-axis according to their genomic positions on the x-axis. POLE ECCs were found to harbor “flat” copy number profiles devoid of gene copy number alterations (top), MMR-D ECCs and a subset of p53 wild-type ECCs harbored few gene copy number alterations (middle), in contrast to ECCs of p53 abnormal subtype (bottom). Morphologic features were not predictive of molecular subtype. |
path4947-sup-0005-FigureS4.tifTIFF image, 451.7 KB | Figure S4. Hierarchical clustering of endometrial clear cell carcinomas from this study and endometrioid and serous carcinomas from TCGA using somatic mutations identified in 300 cancer genes. Hierarchical cluster analysis of mutations identified in the 300 cancer genes included in our targeted massively parallel sequencing assay using Euclidean distance metric and Ward's algorithm, including all endometrial clear cell carcinomas (ECCs) from the current study, and all endometrial endometrioid and serous carcinomas from TCGA. The sample names are shown below the phenobar and the gene names to the right of the heatmap. The tumor type as well as the genomic subtype of the endometrial endometrioid and serous carcinomas as defined by TOGA are presented in the phenobar below the heatmap, color-coded according to the legend. |
path4947-sup-0006-FigureS5.tifTIFF image, 1.9 MB | Figure S5. Assessment of the stability of the hierarchical cluster analysis. Hierarchical clustering of mutations identified in the 300 cancer genes included in our targeted massively parallel sequencing assay was performed using pcvlust, employing Euclidean distance and Ward's algorithm. Values on the edges of the clustering are p-values (%), and red values are Approximately Unbiased (AU) p-values, and green values are Bootstrap Probability (BP) values. |
path4947-sup-0007-FigureS6.tifTIFF image, 586.7 KB | Figure S6. Copy number profiles of endometrial clear cell carcinomas classified as of copy-number low (endometrioid)/ p53 wild-type subtype using a surrogate model. Representative chromosome plots of endometrial clear cell carcinomas classified as of copy-number low (endometrioid)/ p53 wild-type subtype using a surrogate model [38] showing aberrant gene copy number profiles. Loge ratios are plotted on the y-axis according to their genomic positions indicated on the x-axis. |
path4947-sup-0008-TableS1.pdfPDF document, 390.4 KB | Table S1. List of 300 genes included in the targeted capture massively parallel sequencing assay and frequency of mutations affecting these genes in endometrial clear cell carcinomas from this study and endometrial endometrioid and endometrial serous carcinomas from The Cancer Genome Atlas (TCGA) dataset |
path4947-sup-0009-TableS2.pdfPDF document, 90.7 KB | Table S2. Sequencing statistics of endometrial clear cell carcinomas subjected to targeted massively parallel sequencing |
path4947-sup-0010-TableS3.pdfPDF document, 387.6 KB | Table S3. Somatic mutations identified in endometrial clear cell carcinomas by targeted massively parallel sequencing |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1Creasman WT, Odicino F, Maisonneuve P, et al. Carcinoma of the corpus uteri. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. Int J Gynaecol Obstet 2006; 95(suppl 1): S105–S143.
- 2Zaino R, Carinelli SG, Ellenson LH, et al. Epithelial tumours and precursors. In World Health Organization Classification of Tumours of Female Reproductive Organs, RJ Kurman, M-L Carcangiu, CS Herrington, et al. (eds). IARC Pres: Lyon, 2014; 125–134.
- 3Fadare O, Zheng W, Crispens MA, et al. Morphologic and other clinicopathologic features of endometrial clear cell carcinoma: a comprehensive analysis of 50 rigorously classified cases. Am J Cancer Res 2013; 3: 70–95.
- 4Han G, Soslow RA, Wethington S, et al. Endometrial carcinomas with clear cells: a study of a heterogeneous group of tumors including interobserver variability, mutation analysis, and immunohistochemistry with HNF-1beta. Int J Gynecol Pathol 2015; 34: 323–333.
- 5DeLair D, Oliva E, Kobel M, et al. Morphologic spectrum of immunohistochemically characterized clear cell carcinoma of the ovary: a study of 155 cases. Am J Surg Pathol 2011; 35: 36–44.
- 6Fadare O, Parkash V, Dupont WD, et al. The diagnosis of endometrial carcinomas with clear cells by gynecologic pathologists: an assessment of interobserver variability and associated morphologic features. Am J Surg Pathol 2012; 36: 1107–1118.
- 7Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol 1983; 15: 10–17.
- 8Murali R, Soslow RA, Weigelt B. Classification of endometrial carcinoma: more than two types. Lancet Oncol 2014; 15: e268–e278.
- 9Felix AS, Weissfeld JL, Stone RA, et al. Factors associated with Type I and Type II endometrial cancer. Cancer Causes Control 2010; 21: 1851–1856.
- 10Plataniotis G, Castiglione M. Endometrial cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2010; 21(suppl 5): v41–v45.
- 11Cancer Genome Atlas Research Network, Kandoth C, Schultz N, et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013; 497: 67–73.
- 12An HJ, Logani S, Isacson C, et al. Molecular characterization of uterine clear cell carcinoma. Mod Pathol 2004; 17: 530–537.
- 13Hoang LN, McConechy MK, Meng B, et al. Targeted mutation analysis of endometrial clear cell carcinoma. Histopathology 2015; 66: 664–674.
- 14Le Gallo M, Rudd ML, Urick ME, et al. Somatic mutation profiles of clear cell endometrial tumors revealed by whole exome and targeted gene sequencing. Cancer 2017; http://doi.org/10.1002/cncr.30745.
- 15Garg K, Leitao MM, Jr, Kauff ND, et al. Selection of endometrial carcinomas for DNA mismatch repair protein immunohistochemistry using patient age and tumor morphology enhances detection of mismatch repair abnormalities. Am J Surg Pathol 2009; 33: 925–933.
- 16Garg K, Leitao MM, Jr, Wynveen CA, et al. p53 overexpression in morphologically ambiguous endometrial carcinomas correlates with adverse clinical outcomes. Mod Pathol 2010; 23: 80–92.
- 17Ye J, Zhou Y, Weiser MR, et al. Immunohistochemical detection of ARID1A in colorectal carcinoma: loss of staining is associated with sporadic microsatellite unstable tumors with medullary histology and high TNM stage. Hum Pathol 2014; 45: 2430–2436.
- 18Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn 2015; 17: 251–264.
- 19Schultheis AM, Ng CK, De Filippo MR, et al. Massively parallel sequencing-based clonality analysis of synchronous endometrioid endometrial and ovarian carcinomas. J Natl Cancer Inst 2016; 108: djv427.
- 20Li H, Durbin R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 2009; 25: 1754–1760.
- 21DePristo MA, Banks E, Poplin R, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet 2011; 43: 491–498.
- 22Piscuoglio S, Ng CK, Murray MP, et al. The genomic landscape of male breast cancers. Clin Cancer Res 2016; 22: 4045–4056.
- 23Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res 2016; 44: e131.
- 24Cibulskis K, Lawrence MS, Carter SL, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013; 31: 213–219.
- 25Saunders CT, Wong WS, Swamy S, et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 2012; 28: 1811–1817.
- 26Koboldt DC, Zhang Q, Larson DE, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res 2012; 22: 568–576.
- 27De Mattos-Arruda L, Weigelt B, Cortes J, et al. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle. Ann Oncol 2014; 25: 1729–1735.
- 28Chang MT, Asthana S, Gao SP, et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat Biotechnol 2016; 34: 155–163.
- 29Carter SL, Cibulskis K, Helman E, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol 2012; 30: 413–421.
- 30Carter H, Chen S, Isik L, et al. Cancer-specific high-throughput annotation of somatic mutations: computational prediction of driver missense mutations. Cancer Res 2009; 69: 6660–6667.
- 31Choi Y, Chan AP. PROVEAN web server: a tool to predict the functional effect of amino acid substitutions and indels. Bioinformatics 2015; 31: 2745–2747.
- 32Martelotto LG, Ng CK, De Filippo MR, et al. Benchmarking mutation effect prediction algorithms using functionally validated cancer-related missense mutations. Genome Biol 2014; 15: 484.
- 33Shihab HA, Gough J, Cooper DN, et al. Predicting the functional consequences of cancer-associated amino acid substitutions. Bioinformatics 2013; 29: 1504–1510.
- 34Schwarz JM, Rodelsperger C, Schuelke M, et al. MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods 2010; 7: 575–576.
- 35Futreal PA, Coin L, Marshall M, et al. A census of human cancer genes. Nat Rev Cancer 2004; 4: 177–183.
- 36Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and significance across 12 major cancer types. Nature 2013; 502: 333–339.
- 37Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 2014; 505: 495–501.
- 38Talhouk A, McConechy MK, Leung S, et al. A clinically applicable molecular-based classification for endometrial cancers. Br J Cancer 2015; 113: 299–310.
- 39Suzuki R, Shimodaira H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics 2006; 22: 1540–1542.
- 40Lax SF, Pizer ES, Ronnett BM, et al. Clear cell carcinoma of the endometrium is characterized by a distinctive profile of p53, Ki-67, estrogen, and progesterone receptor expression. Hum Pathol 1998; 29: 551–558.
- 41Billingsley CC, Cohn DE, Mutch DG, et al. Prognostic significance of POLE exonuclease domain mutations in high-grade endometrioid endometrial cancer on survival and recurrence: a subanalysis. Int J Gynecol Cancer 2016; 26: 933–938.
- 42Church DN, Stelloo E, Nout RA, et al. Prognostic significance of POLE proofreading mutations in endometrial cancer. J Natl Cancer Inst 2015; 107: 402.
- 43Hussein YR, Weigelt B, Levine DA, et al. Clinicopathological analysis of endometrial carcinomas harboring somatic POLE exonuclease domain mutations. Mod Pathol 2015; 28: 505–514.
- 44Mills AM, Liou S, Ford JM, et al. Lynch syndrome screening should be considered for all patients with newly diagnosed endometrial cancer. Am J Surg Pathol 2014; 38: 1501–1509.
- 45Kobel M, Tessier-Cloutier B, Leo J, et al. Frequent mismatch repair protein deficiency in mixed endometrioid and clear cell carcinoma of the endometrium. Int J Gynecol Pathol 2017; http://doi.org/10.1097/PGP.0000000000000369.
- 46Hall PA, McCluggage WG. Assessing p53 in clinical contexts: unlearned lessons and new perspectives. J Pathol 2006; 208: 1–6.
- 47Schultheis AM, Martelotto LG, De Filippo MR, et al. TP53 mutational spectrum in endometrioid and serous endometrial cancers. Int J Gynecol Pathol 2016; 35: 289–300.
- 48Soussi T, Leroy B, Taschner PE. Recommendations for analyzing and reporting TP53 gene variants in the high-throughput sequencing era. Hum Mutat 2014; 35: 766–778.
- 49Modica I, Soslow RA, Black D, et al. Utility of immunohistochemistry in predicting microsatellite instability in endometrial carcinoma. Am J Surg Pathol 2007; 31: 744–751.
- 50Myers AP. New strategies in endometrial cancer: targeting the PI3K/mTOR pathway – the devil is in the details. Clin Cancer Res 2013; 19: 5264–5274.
- 51Jones S, Wang TL, Shih Ie M, et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 2010; 330: 228–231.
- 52Wiegand KC, Shah SP, Al-Agha OM, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med 2010; 363: 1532–1543.
- 53Bitler BG, Aird KM, Garipov A, et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med 2015; 21: 231–238.
- 54Bitler BG, Fatkhutdinov N, Zhang R. Potential therapeutic targets in ARID1A-mutated cancers. Expert Opin Ther Targets 2015; 19: 1419–1422.
- 55Piulats JM, Matias-Guiu X. Immunotherapy in endometrial cancer: in the nick of time. Clin Cancer Res 2016; 22: 5623–5625.