Cancer-Associated Mutations in Endometriosis without Cancer

Michael S Anglesio, Nickolas Papadopoulos, Ayse Ayhan, Tayyebeh M Nazeran, Michaël Noë, Hugo M Horlings, Amy Lum, Siân Jones, Janine Senz, Tamer Seckin, Julie Ho, Ren-Chin Wu, Vivian Lac, Hiroshi Ogawa, Basile Tessier-Cloutier, Rami Alhassan, Amy Wang, Yuxuan Wang, Joshua D Cohen, Fontayne Wong, Adnan Hasanovic, Natasha Orr, Ming Zhang, Maria Popoli, Wyatt McMahon, Laura D Wood, Austin Mattox, Catherine Allaire, James Segars, Christina Williams, Cristian Tomasetti, Niki Boyd, Kenneth W Kinzler, C Blake Gilks, Luis Diaz, Tian-Li Wang, Bert Vogelstein, Paul J Yong, David G Huntsman, Ie-Ming Shih, Michael S Anglesio, Nickolas Papadopoulos, Ayse Ayhan, Tayyebeh M Nazeran, Michaël Noë, Hugo M Horlings, Amy Lum, Siân Jones, Janine Senz, Tamer Seckin, Julie Ho, Ren-Chin Wu, Vivian Lac, Hiroshi Ogawa, Basile Tessier-Cloutier, Rami Alhassan, Amy Wang, Yuxuan Wang, Joshua D Cohen, Fontayne Wong, Adnan Hasanovic, Natasha Orr, Ming Zhang, Maria Popoli, Wyatt McMahon, Laura D Wood, Austin Mattox, Catherine Allaire, James Segars, Christina Williams, Cristian Tomasetti, Niki Boyd, Kenneth W Kinzler, C Blake Gilks, Luis Diaz, Tian-Li Wang, Bert Vogelstein, Paul J Yong, David G Huntsman, Ie-Ming Shih

Abstract

Background: Endometriosis, defined as the presence of ectopic endometrial stroma and epithelium, affects approximately 10% of reproductive-age women and can cause pelvic pain and infertility. Endometriotic lesions are considered to be benign inflammatory lesions but have cancerlike features such as local invasion and resistance to apoptosis.

Methods: We analyzed deeply infiltrating endometriotic lesions from 27 patients by means of exomewide sequencing (24 patients) or cancer-driver targeted sequencing (3 patients). Mutations were validated with the use of digital genomic methods in microdissected epithelium and stroma. Epithelial and stromal components of lesions from an additional 12 patients were analyzed by means of a droplet digital polymerase-chain-reaction (PCR) assay for recurrent activating KRAS mutations.

Results: Exome sequencing revealed somatic mutations in 19 of 24 patients (79%). Five patients harbored known cancer driver mutations in ARID1A, PIK3CA, KRAS, or PPP2R1A, which were validated by Safe-Sequencing System or immunohistochemical analysis. The likelihood of driver genes being affected at this rate in the absence of selection was estimated at P=0.001 (binomial test). Targeted sequencing and a droplet digital PCR assay identified KRAS mutations in 2 of 3 patients and 3 of 12 patients, respectively, with mutations in the epithelium but not the stroma. One patient harbored two different KRAS mutations, c.35G→T and c.35G→C, and another carried identical KRAS c.35G→A mutations in three distinct lesions.

Conclusions: We found that lesions in deep infiltrating endometriosis, which are associated with virtually no risk of malignant transformation, harbor somatic cancer driver mutations. Ten of 39 deep infiltrating lesions (26%) carried driver mutations; all the tested somatic mutations appeared to be confined to the epithelial compartment of endometriotic lesions.

Figures

Figure 1. An Example of Deeply Infiltrative…
Figure 1. An Example of Deeply Infiltrative Endometriosis in the Colon
In Panel A, a segment of involved colon shows a papillary lesion projecting into the lumen. Panel B shows the cross section of the colonic wall that is indicated by the rectangle in Panel A; arrows indicate endometriotic lesions. In Panel C, a section stained with hematoxylin and eosin shows multiple, discrete endometriotic foci (arrows) infiltrating into the muscle layer of the colon. The box indicates the approximate region that is core-targeted for molecular analysis. In Panel D, a higher magnification shows the characteristic morphologic features of endometriosis, with both glandular and stromal components. Results for all 24 exome-sequenced patients are shown in Panel E; not all patients’ samples yielded detectable somatic mutations. Both “driver” and “passenger” mutations are indicated (Tables S1 and S2 in Supplementary Appendix 2). As opposed to driver mutations, passenger mutations are defined as somatic mutations that are not known or presumed to directly contribute to cancer initiation or progression.
Figure 2. Immunohistochemistry of ARID1A (BAF250A) in…
Figure 2. Immunohistochemistry of ARID1A (BAF250A) in Deep Infiltrating Endometriosis
ARID1A immunoreactivity was detected in all stromal (St) cells and epithelial (Epi) cells within an endometriotic lesion containing wild-type ARID1A (Panel A). In Patient 20, harboring an ARID1A inactivating mutation, loss of ARID1A immunoreactivity was observed in a subset of epithelial cells (arrows indicate examples), but immunoreactivity was preserved in a much larger fraction of the adjacent stromal cells within the same lesion (Panel B). The mutant-allele fraction of the ARID1A mutation in this patient was 8%.
Figure 3. Confirmation of Activating, Somatic KRAS…
Figure 3. Confirmation of Activating, Somatic KRAS Mutations in the Glandular Epithelium but not Stromal Compartments of Deep Endometriotic Lesions
Panel A shows a photomicrograph of endometriotic tissue from Patient 25, with standard hematoxylin and eosin staining. Panels B and C show manually stained, non–cover-slipped sections, also from Patient 25, that were prepared for laser-capture microdissection. Panels D and E show droplet digital polymerase-chain- reaction (PCR) plots illustrating the presence of both c.35G→T (p.G12V) and c.35G→C (p.G12A) KRAS mutations at different allelic frequencies but exclusive to the glandular epithelium in Patient 25. The allelic frequencies represent the percentage of droplets that were positive for the mutant allele (mut+) or positive for the wild-type allele (wt+). Control cell lines and no-template controls (including all reaction components except a DNA template) are also shown. Comp-PMT1 denotes spectrally compensated photomultiplier tube 1 (the dye channel used for the mutant assay), and Comp-PMT2 spectrally compensated photo-multiplier tube 2 (the dye channel used for the wild-type assay).
Figure 4. Co-Occurring and Anatomically Distinct Deep…
Figure 4. Co-Occurring and Anatomically Distinct Deep Infiltrating Endometriosis Lesions That Harbor Identical KRAS Mutations
Panel A shows an anatomical diagram outlining the position of three distinct deep infiltrating endometriosis lesions (in red) and normal sampling of eutopic endometrial and endocervical epithelium (in gray), all from Patient 28. The allelic frequency of the epithelial-restricted c.35G→A (p.G12D) KRAS mutation from droplet digital PCR experiments is also shown below each block identifier. Panel B shows droplet digital PCR plots confirming the KRAS mutation in the epithelial, but not stromal, component of the A8 endometriotic lesion, with the mutant-droplet-positive fraction (allelic frequency). Controls are also shown. Panel C shows hematoxylin-and-eosin photomicrographs of each location shown in Panel A, including endometriotic tissue taken from the three distinct lesions: in the anterior serosal surface of the uterus (A8) as well as the vaginal (B3) and rectal (C2) surfaces of the rectouterine pouch.

References

    1. Giudice LC. Endometriosis. N Engl J Med. 2010;362:2389–98.
    1. Reese KA, Reddy S, Rock JA. Endometriosis in an adolescent population: the Emory experience. J Pediatr Adolesc Gynecol. 1996;9:125–8.
    1. Bulun SE. Endometriosis. N Engl J Med. 2009;360:268–79.
    1. Leyland N, Casper R, Laberge P, Singh SS. Endometriosis: diagnosis and management. J Obstet Gynaecol Can. 2010;32(Suppl 2):S1–S32.
    1. Tosti C, Pinzauti S, Santulli P, Chapron C, Petraglia F. Pathogenetic mechanisms of deep infiltrating endometriosis. Reprod Sci. 2015;22:1053–9.
    1. Ferrero S, Alessandri F, Racca A, Leone Roberti Maggiore U. Treatment of pain associated with deep endometriosis: alternatives and evidence. Fertil Steril. 2015;104:771–92.
    1. Donnez J, Squifflet J, Pirard C, Jadoul P, Wyns C, Smets M. The efficacy of medical and surgical treatment of endometriosis-associated infertility and pelvic pain. Gynecol Obstet Invest. 2002;54(Suppl 1):2–7.
    1. Ferguson BR, Bennington JL, Haber SL. Histochemistry of mucosubstances and histology of mixed Müllerian pelvic lymph node glandular inclusions: evidence for histogenesis by Müllerian metaplasia of coelomic epithelium. Obstet Gynecol. 1969;33:617–25.
    1. Sampson JA. Metastatic or embolic endometriosis, due to the menstrual dissemination of endometrial tissue into the venous circulation. Am J Pathol. 1927;3 93-110.43.
    1. Sampson JA. Peritoneal endometriosis due to menstrual dissemination of endometrial tissue into the peritoneal cavity. Am J Obstet Gynecol. 1927;14:422–69.
    1. Sasson IE, Taylor HS. Stem cells and the pathogenesis of endometriosis. Ann N Y Acad Sci. 2008;1127:106–15.
    1. Figueira PG, Abrão MS, Krikun G, Taylor HS. Stem cells in endometrium and their role in the pathogenesis of endometriosis. Ann N Y Acad Sci. 2011;1221:10–7.
    1. Fung JN, Rogers PA, Montgomery GW. Identifying the biological basis of GWAS hits for endometriosis. Biol Reprod. 2015;92:87.
    1. Anglesio MS, Bashashati A, Wang YK, et al. Multifocal endometriotic lesions associated with cancer are clonal and carry a high mutation burden. J Pathol. 2015;236:201–9.
    1. Kurman RJ, Shih IM. The dualistic model of ovarian carcinogenesis: revisited, revised, and expanded. Am J Pathol. 2016;186:733–47.
    1. Vestergaard AL, Thorup K, Knudsen UB, et al. Oncogenic events associated with endometrial and ovarian cancers are rare in endometriosis. Mol Hum Reprod. 2011;17:758–61.
    1. Sato N, Tsunoda H, Nishida M, et al. Loss of heterozygosity on 10q23.3 and mutation of the tumor suppressor gene PTEN in benign endometrial cyst of the ovary: possible sequence progression from benign endometrial cyst to endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer Res. 2000;60:7052–6.
    1. Samartzis EP, Samartzis N, Noske A, et al. Loss of ARID1A/BAF250a–expression in endometriosis: a biomarker for risk of carcinogenic transformation? Mod Pathol. 2012;25:885–92.
    1. Borrelli GM, Abrão MS, Taube ET, et al. (Partial) loss of BAF250a (ARID1A) in recto-vaginal deep-infiltrating endometriosis, endometriomas and involved pelvic sentinel lymph nodes. Mol Hum Reprod. 2016;22:329–37.
    1. Chene G, Ouellet V, Rahimi K, Barres V, Provencher D, Mes-Masson AM. The ARID1A pathway in ovarian clear cell and endometrioid carcinoma, contiguous endometriosis, and benign endometriosis. Int J Gynaecol Obstet. 2015;130:27–30.
    1. Fassbender A, Rahmioglu N, Vitonis AF, et al. World Endometriosis Research Foundation Endometriosis Phenome, Biobanking Harmonisation Project: IV. Tissue collection, processing, and storage in endometriosis research. Fertil Steril. 2014;102:1244–53.
    1. Robles AI, Traverso G, Zhang M, et al. Whole-exome sequencing analyses of inflammatory bowel disease-associated colorectal cancers. Gastroenterology. 2016;150:931–43.
    1. Anglesio MS, Wang YK, Maassen M, et al. Synchronous endometrial and ovarian carcinomas: evidence of clonality. J Natl Cancer Inst. 2016;108(6):djv428.
    1. Kinde I, Wu J, Papadopoulos N, Kinzler KW, Vogelstein B. Detection and quantification of rare mutations with massively parallel sequencing. Proc Natl Acad Sci U S A. 2011;108:9530–5.
    1. Ayhan A, Mao TL, Seckin T, et al. Loss of ARID1A expression is an early molecular event in tumor progression from ovarian endometriotic cyst to clear cell and endometrioid carcinoma. Int J Gynecol Cancer. 2012;22:1310–5.
    1. Guan B, Mao TL, Panuganti PK, et al. Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. Am J Surg Pathol. 2011;35:625–32.
    1. Wu RC, Ayhan A, Maeda D, et al. Frequent somatic mutations of the telomerease reverse transcriptase promoter in ovarian clear cell carcinoma but not in other major types of gynaecological malignancy. J Pathol. 2014;232:473–81.
    1. Mao TL, Ardighieri L, Ayhan A, et al. Loss of ARID1A expression correlates with stages of tumor progression in uterine endometrioid carcinoma. Am J Surg Pathol. 2013;37:1342–8.
    1. De Graaff AA, D’Hooghe TM, Dunselman GA, Dirksen CD, Hummelshoj L, Simoens S. The significant effect of Endometriosis on physical, mental and social wellbeing: results from an international cross-sectional survey. Hum Reprod. 2013;28:2677–85.
    1. Cullen TS. Adenoma-myoma uteri diffusum benignum. Johns Hopkins Hosp Bull. 1896;6:133–7.
    1. Russell WW. Aberrant portions of the Mullerian duct found in an ovary. Johns Hopkins Hosp Bull. 1899;10:8–9.
    1. Gao X, Outley J, Botteman M, Spalding J, Simon JA, Pashos CL. Economic burden of endometriosis. Fertil Steril. 2006;86:1561–72.
    1. Klein S, D’Hooghe T, Meuleman C, Dirksen C, Dunselman G, Simoens S. What is the societal burden of endometriosis-associated symptoms? A prospective Belgian study. Reprod Biomed Online. 2014;28:116–24.
    1. Simoens S, Dunselman G, Dirksen C, et al. The burden of endometriosis: costs and quality of life of women with endometriosis and treated in referral centres. Hum Reprod. 2012;27:1292–9.
    1. McConechy MK, Anglesio MS, Kalloger SE, et al. Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas. J Pathol. 2011;223:567–73.
    1. Stratton MR, Campbell PJ, Futreal PA. The cancer genome. Nature. 2009;458:719–24.
    1. Forbes SA, Bindal N, Bamford S, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2011;39:D945–D950.
    1. Wiegand KC, Shah SP, Al-Agha OM, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 2010;363:1532–43.
    1. Thomas EJ, Campbell IG. Molecular genetic defects in endometriosis. Gynecol Obstet Invest. 2000;50(Suppl 1):44–50.
    1. Yamamoto S, Tsuda H, Takano M, Tamai S, Matsubara O. Loss of ARID1A protein expression occurs as an early event in ovarian clear-cell carcinoma development and frequently coexists with PIK3CA mutations. Mod Pathol. 2012;25:615–24.
    1. Wei JJ, William J, Bulun S. Endometriosis and ovarian cancer: a review of clinical, pathologic, and molecular aspects. Int J Gynecol Pathol. 2011;30:553–68.
    1. Cheng CW, Licence D, Cook E, et al. Activation of mutated Kras in donor endometrial epithelium and stroma promotes lesion growth in an intact immunocompetent murine model of endometriosis. J Pathol. 2011;224:261–9.
    1. Martincorena I, Roshan A, Gerstung M, et al. Tumor evolution: high burden and pervasive positive selection of somatic mutations in normal human skin. Science. 2015;348:880–6.
    1. McConnell MJ, Lindberg MR, Brennand KJ, et al. Mosaic copy number variation in human neurons. Science. 2013;342:632–7.
    1. Krimmel JD, Schmitt MW, Harrell MI, et al. Ultra-deep sequencing detects ovarian cancer cells in peritoneal fluid and reveals somatic TP53 mutations in non-cancerous tissues. Proc Natl Acad Sci U S A. 2016;113:6005–10.
    1. Kato S, Lippman SM, Flaherty KT, Kurzrock R. The conundrum of genetic “drivers” in benign conditions. J Natl Cancer Inst. 2016;108:108.
    1. Masuda H, Matsuzaki Y, Hiratsu E, et al. Stem cell-like properties of the endometrial side population: implication in endometrial regeneration. PLoS One. 2010;5(4):e10387.
    1. Cervelló I, Mas A, Gil-Sanchis C, et al. Reconstruction of endometrium from human endometrial side population cell lines. PLoS One. 2011;6(6):e21221.
    1. Jubanyik KJ, Comite F. Extrapelvic endometriosis. Obstet Gynecol Clin North Am. 1997;24:411–40.
    1. Vercellini P, Fedele L, Aimi G, Pietropaolo G, Consonni D, Crosignani PG. Association between endometriosis stage, lesion type, patient characteristics and severity of pelvic pain symptoms: a multivariate analysis of over 1000 patients. Hum Reprod. 2007;22:266–71.

Source: PubMed

3
Abonner