1,373
Views
13
CrossRef citations to date
0
Altmetric
Editorial

Identifying aggressive forms of endometrioid-type endometrial cancer: new insights into molecular subtyping

, &

Abstract

Clinical heterogeneity represents a great challenge for cancer therapeutics. Molecular classification of patients into different subtypes based on genetic or epigenetic characteristics has the potential to revolutionize the clinical care and mechanistic understanding of a wide spectrum of cancers, including endometrial carcinoma, the most common gynecological cancer affecting women.

Clinical heterogeneity and complexity represent a great challenge for cancer therapeutics. Cancer classification based primarily on microscopic appearance of the tumor, such as stage, grade or cell lineage has allowed for some successes in individualizing cancer therapy. Microscopic morphology, however, has its limitations when projecting treatment strategies and cancer clinical course. Consequently, oncologists have begun to incorporate genetic classifications to guide clinical decision making Citation[1]. This approach has revolutionized the clinical care and mechanistic understanding of different cancers. Breast cancer patients are clinically categorized into three intrinsic gene expression subtypes with different therapeutic options Citation[2]. Endocrine therapy is provided to the estrogen receptor (ER) positive group, the HER2/ERBB2 amplified group receives Herceptin therapy, and triple negative breast cancers are treated with chemotherapy. Mutations in IDH1 or IDH2 define a subtype of diffuse lower grade gliomas with favorable prognosis Citation[3]. The Cancer Genome Atlas (TCGA), a collaborative project of the National Cancer Institute and the National Human Genome Research Institute, has recently generated catalogs of multi-dimensional molecular abnormalities in a large population of patient samples in numerous cancer types Citation[4]. In-depth bioinformatic and statistical analysis of the TCGA datasets has revealed new molecular subtypes that have potential clinical implications. A Glioma CpG island methylator phenotype that was characteristic of CpG island methylation and associated with favorable prognosis wass recently identified using the TCGA glioblastoma multiforme datasets Citation[5]. Genetic or epigenetic biomarkers are increasingly used in clinical practice to stratify patients based on difference in survival or response to chemotherapy. High-grade serous ovarian carcinomas with BRCA2 and, to a lesser extent, BRCA1 mutations are deficient in the induction of homologous recombination and therefore have an improved sensitivity to chemotherapy and better survival Citation[6].

Endometrial carcinoma is the fourth most common cancer among women after breast, lung and colorectal cancer. Approximately 49,560 uterine corpus cases are estimated to be diagnosed in the USA in 2013 Citation[7]. There are two broad microscopic subtypes, endometrioid and non-endometrioid. Recognizing these microscopic subtypes is important, as the non-endometrioid carcinomas are typically more clinically aggressive, presenting with advanced stage at the time of diagnosis. Not surprisingly, these two histologies of endometrial carcinoma have distinct molecular aberrations Citation[8]. Molecular clustering of all endometrial carcinoma samples together commonly segregates cases based largely on differences between serous versus endometrioid histologies Citation[8,9]. Endometrioid-type endometrial carcinoma (EEC) accounts for approximately 70–80% of all endometrial cancers, but it is less characterized at the genomic level, especially in large patient cohorts. The two isoforms of ER (ERα and ERβ), encoded by ESR1 and ESR2, respectively, have different functions in cancer biology and therapy Citation[10]. In particular, ERα, the predominant isoform in EEC, predicts response to endocrine therapy and is correlated with better survival, whereas ERβ did not demonstrate significant correlations with clinicopathological characteristics Citation[10]. Although many patients with EEC are cured by surgery (hysterectomy) alone without chemotherapy, it is well known in the oncology community that a subset of these patients will have recurrence. Predicting which patient will have recurrence is currently not possible.

Identification of molecular subtypes of EEC

The clinical heterogeneity of EEC suggests that there are several distinct biological phenotypes that may result from genetic variations that alter oncogenic pathways during tumorigenesis. Recently, we performed an integrated analysis on the multiple-dimensional data types, including whole-exome and RNA sequencing, reverse phase protein array profiling and clinical data from the TCGA database. We restricted the analyses to EEC (n = 271), specifically excluding serous carcinomas, and identified four transcriptome subtypes with distinct clinicopathologic characteristics and mutation spectra Citation[11]. Cluster II (n = 61) consisted of younger, obese EEC patients with low-grade, low-stage disease at the time of initial diagnosis, but exhibiting a diminished survival compared with Cluster I, which was similarly composed of low-grade, low-stage carcinomas but had higher expression of hormone receptors. CTNNB1 exon 3 mutations were present in 87% of the carcinomas in Cluster II. High expression of Wnt/β-catenin signaling downstream targets, such as Cyclin D1, were also associated with significantly poorer overall survival. Interestingly, Cluster II has the lowest overall mutation rate of the four clusters, suggesting that this cluster is driven by CTNNB1 mutation. At about the same time as our TCGA study was being published, Myers et al. demonstrated in a letter to the editor the association of CTNNB1 mutation with an increased risk for disease relapse in patients with International Federation of Gynecology and Obstetrics Stage 1A, grade 1 endometrioid endometrial carcinoma Citation[12]. This provides independent confirmation that CTNNB1 mutation is important prognostically for patients with low-grade, low-stage endometrioid carcinomas. In addition, CTNNB1 alterations are significantly correlated with EEC patients with younger age Citation[11], and have been previously detected in endometrial hyperplasias Citation[13], suggesting that CTNNB1 alteration is an early event and plays an important role in endometrial carcinogenesis.

Clinical insights & implications

The current clinical dogma for endometrial cancer is that younger patients with endometrioid-type carcinomas are more likely obese with hormone-driven tumors that are low grade and low stage at the time of diagnosis and thus have a better prognosis Citation[14]. Our results clearly show that this group of patients is heterogeneous at the molecular and clinical level. Cluster I, characterized by higher levels of hormone receptors, fits the traditional view of obese endometrial cancer patients and exhibited better clinical outcome. However, Cluster II, also consisting of obese patients, is driven by CTNNB1 mutations and consequently exhibited a decreased survival compared with Cluster I. Identification of this Cluster II subtype helps to explain, at least in part, the clinical heterogeneity of EEC.

The CTNNB1 mutations in Cluster II were primarily confined to a small region in exon 3. This mutational hotspot contains the phosphorylation target for the glycogen synthase kinase-3β kinase, inhibiting its ability to phosphorylate β-catenin (encoded by CTNNB1 gene), which results in β-catenin accumulation within the cell nucleus and activation of transcriptional programs. Integrated analysis of gene expression profiling showed that genes positively correlated with CTNNB1 mutation were significantly enriched with components of the Wnt signaling pathway Citation[15]. Using the presence of CTNNB1 exon 3 mutation, patients could be predicted to have a Cluster II genotype with a sensitivity of 87.0% and a specificity of 88.0%. Of note, the CTNNB1 mutation hotspot contained almost one-third non-synonymous mutations, resulting in amino acid substitutions of codons 32, 34 and 35 that are not known to be glycogen synthase kinase-3β phosphorylation targets. The effect of these mutations on the β-catenin protein accumulation in the nucleus is not clear yet and warrants functional investigation.

There are several ways in which this new and exciting data may be clinically translated. Patients with grade 1 or grade 2 EEC may have CTNNB1 sequencing performed using tumor acquired during the endometrial biopsy to establish diagnosis. Current advance in targeted next-generation sequencing makes CTNNB1 hotspot mutation sequencing analysis less expensive and clinically practical. For patients with such mutations, more aggressive therapeutic approaches could be considered, such as more extensive surgery to include lymphadenectomy and even post-operative adjuvant radiation treatment and/or chemotherapy. Until recently, specific therapeutic targeting of the Wnt/β-catenin signaling pathway was not feasible. However, there are now several drugs in early clinical trials. PRI-724 inhibits β-catenin recruitment of its co-activator CREB binding protein, thereby blocking transcription of Wnt pathway genes Citation[16]. LGK974 inhibits the O-acyltransferase porcupine that acylates Wnt proteins for secretion Citation[17]. Cyclin D1 protein interacts with CDK4/6 to promote cell cycle progression. LEE001 is CDK4/6 inhibitor that could potentially benefit patients with CTNNB1 mutant endometrial carcinomas Citation[18]. CTNNB1 genotyping could also provide value in women with endometrial hyperplasia or grade 1 EEC who are being managed conservatively with some type of progesterone preparation, such as the Mirena IUD Citation[19]. It is known that a subset of these patients will not completely respond to such conservative treatment. Based on our previous analyses, it is plausibly speculated that the non-responders may have tumors with CTNNB1 mutation, as these tumors typically have relatively lower expression of the hormone receptors and are not characterized by high expression of estrogen-induced genes. More effective conservative management of these CTNNB1 mutant tumors might be achieved by adding another agent to the progesterone regimen, such as a PI3K/AKT/mTOR inhibitor, as mutations in this signaling pathway are common across all subtypes of EEC. Similar to the agents targeting mutant IDH1 and IDH2 Citation[20], drugs that can directly inhibit mutant CTNNB1 can be developed in the future and will certainly benefit the EEC patients with this mutation. In summary, the identification of this second cluster of patients with endometrial cancer helps to refute the long-standing knowledge that young, obese patients universally have endometrial cancers that are estrogen-driven and thus have a good prognosis and may help pave the way for more individualized therapy of endometrial cancer patients.

Financial & competing interests disclosure

This study was partially supported by a Career Development Award from MD Anderson Gynecologic SPORE in Uterine Cancers (NIH 2P50 CA098258-08 to Y Liu), funding for the Genome Data Analysis Centers from the National Institutes of Health (U24 CA143835 to W Zhang), and funding for the Cancer Systems Informatics Center from the National Foundation for Cancer Research (W Zhang). The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

References

  • Zhang W, Cheng SY, Hou LF, et al. Genomics in personalized cancer medicine and its impact on early drug development in China: report from the 6th Annual Meeting of the US Chinese Anti-Cancer Association (USCACA) at the 50th ASCO Annual Meeting. Chin J Cancer 2014;33(8):371-5
  • Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumors. Nature 2012;490(7418):61-70
  • Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med 2009;360(8):765-73
  • Zhang W. TCGA divides gastric cancer into four molecular subtypes: implications for individualized therapeutics. Chin J Cancer 2014;33(10):469-70
  • TCGA Research Network. The somatic genomic landscape of glioblastoma. Cell 2013;155(2):462-77
  • Yang D, Khan S, Sun Y, et al. Association of BRCA1 and BRCA2 mutations with survival, chemotherapy sensitivity, and gene mutator phenotype in patients with ovarian cancer. JAMA 2011;306(14):1557-65
  • Siegel R, Naishadham D, Jemal A. Cancer statistics, 2013. CA Cancer J Clin 2013;63(1):11-30
  • Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 2013;497(7447):67-73
  • Salvesen HB, Carter SL, Mannelqvist M, 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(12):4834-9
  • Thomas C, Gustafsson JA. The different roles of ER subtypes in cancer biology and therapy. Nat Rev Cancer 2011;11(8):597-608
  • Liu YX, Patel L, Mills GB, et al. Clinical significance of CTNNB1 mutation and Wnt pathway activation in endometrioid endometrial carcinoma. J Natl Cancer Inst 2014;106(8; dju245
  • Myers A, Barry WT, Hirsch MS, et al. beta-Catenin mutation in recurrent FIGO IA grade 1 endometrioid endometrial cancers. Gynecol Oncol 2014;134(2):426-7
  • Matias-Guiu X, Prat J. Molecular pathology of endometrial carcinoma. Histopathology 2013;62(1):111-23
  • Mhawech-Fauceglia P, Wang D, Samrao D, et al. Trefoil factor family 3 (TFF3) expression and its interaction with estrogen receptor (ER) in endometrial adenocarcinoma. Gynecol Oncol 2013;130(1):174-80
  • Liu C, Li Y, Semenov M, et al. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell 2002;108(6):837-47
  • NCI. Available from: www.cancer.gov/drugdictionary?cdrid=696436
  • Liu J, Pan S, Hsieh MH, et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc Natl Acad Sci USA 2013;110(50):20224-9
  • Dickson MA. Molecular pathways: CDK4 inhibitors for cancer therapy. Clin Cancer Res 2014;20(13):3379-83
  • Ramirez PT, Frumovitz M, Bodurka DC, et al. Hormonal therapy for the management of grade 1 endometrial adenocarcinoma: a literature review. Gynecol Oncol 2004;95(1):133-8
  • Feliciano P. Inhibitors of mutant IDH1 and IDH2. Nat Genet 2013;45(5):477

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.