1,559
Views
13
CrossRef citations to date
0
Altmetric
Editorial

Novel genetic targets in endometrial cancer

Abstract

Worldwide, ∼ 74,000 women die from endometrial cancer each year. Understanding the somatic genomic alterations that drive endometrial tumorigenesis may provide new opportunities to identify targeted therapies for specific subsets of patients. Since 2012, the use of next-generation sequencing to decode the mutational landscape of endometrial tumors has not only confirmed prior knowledge of established genetic targets for serous and endometrioid endometrial carcinomas (ECs), but has also uncovered novel significantly mutated genes, referred to herein as novel genetic targets, which represent candidate cancer genes in these tumors. This editorial summarizes the novel genetic targets that have been identified in serous and endometrioid ECs, according to their unifying functional characteristics. An expert opinion section comments on remaining knowledge gaps that will undoubtedly be filled in future genomic studies of endometrial cancer.

1. Introduction

Most endometrial cancers are endometrial carcinomas (ECs), which are further classified into numerous histopathological subtypes including endometrioid and serous ECs Citation[1]. Endometrioid tumors account for ∼ 80% of newly diagnosed ECs. Although they have a good overall prognosis, improved therapeutic strategies are needed to treat recurrent and advanced-stage endometrioid tumors. Likewise, alternative therapeutic approaches are needed for the treatment of serous ECs, which have a poor overall prognosis. In the era of precision medicine, comprehensive interrogations of tumor exomes and genomes for somatic alterations have been driven by the hope that they will reveal novel genetic targets that might ultimately guide treatment. For the purposes of this editorial, the consideration of novel genetic targets is limited to mutated genes; other classes of genomic and epigenomic alterations also represent novel genetic targets but are beyond the scope of this discussion.

2. Novel genetic targets in endometrioid and serous ECs

Recent massively parallel sequencing of endometrioid and serous ECs by individual laboratories Citation[2-6], and by The Cancer Genome Atlas (TCGA) Citation[7], has uncovered novel, statistically significantly mutated genes (SMGs) in these tumors, that is, genes that are mutated at a statistically significantly higher rate than background and which therefore represent candidate cancer genes. Furthermore, the integrated genomic analysis of endometrioid and serous ECs by TCGA revealed that they can be reclassified into four distinct molecular subgroups: ultramutated/POLE mutant, hypermutated/microsatellite instability (MSI), copy number low/microsatellite stable (MSS) and copy number high/serous-like Citation[7]. The abundance of SMGs in the ultramutated subgroup precludes their discussion in this editorial. Among the remaining 3 subgroups, 32 SMGs, including 20 novel genetic targets (), have been described Citation[7]. For ease of discussion herein, these novel genetic targets are loosely categorized according to their major ascribed functions.

Table 1. Mutation frequency of novel significantly mutated protein-encoding genes identified among serous and non-ultramutated endometrioid ECs.

2.1 Transcriptional regulation

Six novel SMGs, CTCF, ZFHX3 (Zinc finger homeobox 3), SOX17, BCOR, MECOM and TAF1, encode transcriptional regulators. CTCF (CCCTC-binding factor) encodes a zinc-finger transcription factor that regulates transcriptional activation and repression, and influences chromatin architecture Citation[8]. Frequent truncating mutations within CTCF in MSI+ endometrioid ECs, and the unstable nature of the truncated transcripts, have led to the proposal that CTCF may be a haploinsufficient tumor suppressor gene in these tumors Citation[9]. Another transcription factor gene, ZFHX3, is significantly mutated in hypermutated/MSI ECs Citation[7]. ZFHX3 is a putative tumor suppressor gene based on its location within a common region of 16q22 deletion in human cancer and the occurrence of frequent somatic mutations in prostate cancer Citation[10]. SOX17 (SRY [sex determining region Y]-box 17), an SMG in copy number low/MSS ECs, encodes an high mobility group box transcription factor that, among other effects, negatively regulates WNT/β-catenin signaling Citation[11]. In copy number low/MSS ECs, SOX17 mutations are mutually exclusive with other genetic aberrations affecting the WNT/β-catenin pathway, suggesting that they likely perturb WNT signaling Citation[7]. BCOR (BCL6 corepressor), which encodes a corepressor of BCL6, is significantly mutated in copy number low/MSS ECs Citation[7]. MECOM (MDS1 and EVI1 complex locus), a protooncogene that encodes a zinc-finger transcription factor, is significantly mutated in copy number low/MSS ECs Citation[7]. Although the functional impact of MECOM mutations in EC is unknown, it is noteworthy that MECOM inhibitors are being developed Citation[12]. Finally, TAF1 (TAF1 RNA polymerase II, TATA box binding protein-associated factor, 250 kDa) encodes a subunit of the TFIID basal transcription factor and is an SMG in serous EC Citation[4].

2.2 Chromatin remodeling and chromatin organization

CHD4 (chromodomain helicase DNA binding protein 4) and ARID5B (AT-rich interactive domain 5B [MRF1-like]) are novel SMGs involved in chromatin remodeling Citation[3,7]. CHD4 was first implicated in EC by virtue of its designation as an SMG in serous tumors Citation[3]. CHD4 is a subunit of the NuRD complex, which is a transcriptional activator and repressor, and is implicated in the DNA damage response Citation[13]. ARID5B (AT-rich interactive domain 5B [MRF1-like]) is an SMG in hypermutated/MSI ECs and encodes a DNA-binding protein that complexes with PHF2 (PHD finger protein 2), a lysine demethylase. The DNA-binding activity of ARID5B is believed to direct the ARID5B–PHF2 complex to specific gene promoters resulting in histone demethylation and transcriptional activation Citation[14]. Another SMG in hypermutated/MSI ECs is HIST1H2BD (histone cluster 1, H2bd), which encodes a core component of the nucleosome.

2.3 Ubiquitin-mediated protein degradation

FBXW7 (F-Box and WD repeat domain containing 7, E3 ubiquitin protein ligase) and SPOP (speckle-type POZ protein), which encode the substrate recognition components of the SKP1-CUL1-FBXW7 and SPOP-CUL3 ubiquitin ligase complexes, respectively, are SMGs in serous ECs Citation[2-4]. FBXW7 is a bona fide tumor suppressor and SPOP is a putative tumor suppressor. Many of the FBXW7 mutations documented in EC encode dominant-negative or loss-of -function mutants in other tumor types and it is therefore anticipated that SPOP mutations in EC may function similarly Citation[3]. In certain cellular contexts, loss of FBXW7 function is associated with in vitro sensitivity to sorafenib and resistance to antitubulin chemotherapeutics Citation[15], or sensitivity to an histone deacetylase inhibitor Citation[16]. However, the relevance of those observations to FBXW7-mutant endometrial cancers remains to be elucidated.

2.4 RNA binding or RNA modification

Four novel genetic targets, RPL22, RBMX, CSDE1 and METTL14, have RNA-binding capability or RNA modification activity. RPL22 (ribosomal protein L22) encodes a ribosomal protein and is believed to be a tumor suppressor gene in T-ALL Citation[17]. RPL22 mutations are extraordinarily frequent (52%) in MSI+ EECs, and are attributed to a recurrent frameshift mutation (c.43delA) suggesting loss of function Citation[18]. RBMX (RNA-binding motif protein, X-linked) encodes an RNA-binding protein that regulates pre-mRNA splicing Citation[19], and is implicated in the DNA damage response Citation[20], mitotic sister chromatid cohesion Citation[21] and the regulation of tumor necrosis factor receptor 1 release. RBMX is an SMG in hypermutated/MSI ECs, exhibiting recurrent in-frame deletions encompassing the translation start site. CSDE1 (cold shock domain containing E1, RNA-binding), an SMG in hypermutated/MSI EC, encodes an RNA- and single-stranded DNA-binding protein that regulates mRNA turnover and translation Citation[22]. Most CSDE1 mutations in EC are missense mutations, including three recurrently mutated residues (R174, R726, R774). Finally, METTL14 (methyltransferase like 14) is significantly mutated in hypermutated/MSI ECs Citation[7], and encodes a methyltransferase that complexes with METTL3 and methylates m6A on nuclear RNA Citation[23]. The majority of METTL14 mutations in EC are missense mutations within the methyltransferase domain, including a recurrent R298P mutation Citation[7].

2.5 Other functions

NKAP, GIGYF2, LIMCH1, TNFAIP6 and SGK1 are novel genetic targets that do not precisely fit into the previously discussed categories. NKAP (NFKB-activating protein) regulates NF kappa B activation induced by TNF and IL-1 Citation[24], is a transcriptional corepressor for Notch Citation[25] and has been implicated in RNA splicing Citation[26]. NKAP is significantly mutated in hypermutated/MSI ECs Citation[7]. GIGYF2 (GRB10 interacting GYF protein 2), encoded by an SMG in hypermutated/MSI ECs, is involved in IGF-I receptor signaling Citation[27], and is implicated in the regulation of protein translation Citation[28]. LIMCH1 (LIM and Calponin homology domains 1) encodes a protein with a poorly defined function and is significantly mutated in hypermutated/MSI ECs Citation[7]. The R421fs frameshift mutation accounts for almost all LIMCH1 mutations in hypermutated/MSI ECs and is predicted to encode a truncated protein lacking the LIM domain. TNFAIP6 (TNFα-induced protein 6) is significantly mutated in hypermutated/MSI ECs. The encoded protein interacts with components of the extracellular matrix including hyaluronan Citation[29], and is involved in the inflammatory response and tissue remodeling Citation[30]. Finally, SGK1 (serum/glucocorticoid regulated kinase 1), an SMG in copy number low/MSS ECs, encodes a kinase implicated in the regulation of a wide range of cellular processes including tumor cell survival. Although SGK1 has received attention as a potential druggable target in cancer Citation[31], the biological and therapeutic relevance of SGK1 mutations in EC remains unknown.

3. Mutation patterns between SMGs

Patterns of mutations between genes can provide insights into their possible functional effects. Here, the pattern of mutations among the 32 SMGs, identified in non-ultramutated ECs in TCGA, was obtained via the cBioPortal for Cancer Genomics (Supplementary Tables 14) Citation[32,33]. As shown in , mutations in a number of novel SMGs showed a tendency for mutually exclusive or co-occurring mutations, suggesting, respectively, possible functional redundancy or cooperativity.

Table 2. Statistically significant trends in mutation patterns* involving novel SMGs identified in non-ultramutated ECs in the TCGA study Citation[7].

4. Expert opinion

With novel genetic targets for serous and endometrioid EC now in hand, the challenge will be to determine whether any of these targets are therapeutically relevant. This can take the form of biochemical and biological studies of mutant proteins in their proper cellular and genetic context, systems biology approaches to functionalize the genome Citation[5], as well as systematic searches for gene–drug interactions. One strategy to prioritize genetic targets for further analysis is to focus on the most frequently mutated of the SMGs, which, if proven to be druggable, would theoretically maximize the number of patients who might gain clinical benefit. Alternatively, SMGs that have reported gene–drug interactions and/or are being evaluated as druggable targets in preclinical studies within other cellular contexts, such as FBXW7, MECOM and SGK1, could be prioritized for assessment of their therapeutic relevance to EC. In terms of seeking druggable targets, it is noteworthy that catalogs of SMGs are dynamic rather than static and depend on the assumptions and parameters used in statistical calculations as well as user-defined statistical cutoffs. Moreover, current catalogs of genetic targets in serous and endometrioid ECs have principally been derived from analyses of primary tumor tissues resected prior to treatment. As such, there may be additional genetic targets yet to be discovered within recurrent or persistent tumors that are refractory to therapy, or in metastatic disease. Additionally, the genomic landscape of other histological subtypes of EC remains to be elucidated. Finally, it remains to be seen whether additional novel genetic targets will emerge from sequencing of larger numbers of tumors corresponding to each of the four recently defined molecular subgroups of serous and endometrioid EC.

Declaration of interest

D W Bell is a co-inventor on a patent describing EGFR mutations, which is licensed to Genzyme. The author is funded through the Intramural Research Program of the National Human Genome Research Institute at NIH. The author has no other 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 apart from those disclosed.

Supplemental material

Bibliography

  • Dedes KJ, Wetterskog D, Ashworth A, et al. Emerging therapeutic targets in endometrial cancer. Nat Rev Clin Oncol 2011;8:261-71
  • Kuhn E, Wu RC, Guan B, et al. Identification of molecular pathway aberrations in uterine serous carcinoma by genome-wide analyses. J Natl Cancer Inst 2012;104:1503-13
  • Le Gallo M, O'Hara AJ, Rudd ML, et al. Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nat Genet 2012;44:1310-15
  • Zhao S, Choi M, Overton JD, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci USA 2013;110:2916-21
  • Liang H, Cheung LW, Li J, et al. Whole-exome sequencing combined with functional genomics reveals novel candidate driver cancer genes in endometrial cancer. Genome Res 2012;22:2120-9
  • Kinde I, Bettegowda C, Wang Y, et al. Evaluation of DNA from the Papanicolaou test to detect ovarian and endometrial cancers. Sci Transl Med 2013;5:167ra4
  • Kandoth C, Schultz N, Cherniack AD, et al. Integrated genomic characterization of endometrial carcinoma. Nature 2013;497:67-73
  • Lee BK, Iyer VR. Genome-wide studies of CCCTC-binding factor (CTCF) and cohesin provide insight into chromatin structure and regulation. J Biol Chem 2012;287:30906-13
  • Zighelboim I, Mutch DG, Knapp A, et al. High frequency strand slippage mutations in CTCF in MSI-positive endometrial cancers. Hum Mutat 2014;35:63-5
  • Sun X, Frierson HF, Chen C, et al. Frequent somatic mutations of the transcription factor ATBF1 in human prostate cancer. Nat Genet 2005;37:407-12
  • Sinner D, Kordich JJ, Spence JR, et al. Sox17 and Sox4 differentially regulate beta-catenin/T-cell factor activity and proliferation of colon carcinoma cells. Mol Cell Biol 2007;27:7802-15
  • Zhang Y, Sicot G, Cui X, et al. Targeting a DNA binding motif of the EVI1 protein by a pyrrole-imidazole polyamide. Biochemistry 2011;50:10431-41
  • O'Shaughnessy A, Hendrich B. CHD4 in the DNA-damage response and cell cycle progression: not so NuRDy now. Biochem Soc Trans 2013;41:777-82
  • Baba A, Ohtake F, Okuno Y, et al. PKA-dependent regulation of the histone lysine demethylase complex PHF2-ARID5B. Nat Cell Biol 2011;13:668-75
  • Inuzuka H, Fukushima H, Shaik S, et al. Mcl-1 ubiquitination and destruction. Oncotarget 2011;2:239-44
  • Garnett MJ, Edelman EJ, Heidorn SJ, et al. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 2012;483:570-5
  • Rao S, Lee SY, Gutierrez A, et al. Inactivation of ribosomal protein L22 promotes transformation by induction of the stemness factor, Lin28B. Blood 2012;120:3764-73
  • Novetsky AP, Zighelboim I, Thompson DM Jr, et al. Frequent mutations in the RPL22 gene and its clinical and functional implications. Gynecol Oncol 2013;128:470-4
  • Heinrich B, Zhang Z, Raitskin O, et al. Heterogeneous nuclear ribonucleoprotein G regulates splice site selection by binding to CC(A/C)-rich regions in pre-mRNA. J Biol Chem 2009;284:14303-15
  • Adamson B, Smogorzewska A, Sigoillot FD, et al. A genome-wide homologous recombination screen identifies the RNA-binding protein RBMX as a component of the DNA-damage response. Nat Cell Biol 2012;14:318-28
  • Matsunaga S, Takata H, Morimoto A, et al. RBMX: a regulator for maintenance and centromeric protection of sister chromatid cohesion. Cell Rep 2012;1:299-308
  • Mihailovich M, Militti C, Gabaldon T, et al. Eukaryotic cold shock domain proteins: highly versatile regulators of gene expression. Bioessays 2010;32:109-18
  • Liu J, Yue Y, Han D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N(6)-adenosine methylation. Nat Chem Biol 2014;10:93-5
  • Chen D, Li Z, Yang Q, et al. Identification of a nuclear protein that promotes NF-kappaB activation. Biochem Biophys Res Commun 2003;310:720-4
  • Pajerowski AG, Nguyen C, Aghajanian H, et al. NKAP is a transcriptional repressor of notch signaling and is required for T cell development. Immunity 2009;30:696-707
  • Burgute BD, Peche VS, Steckelberg AL, et al. NKAP is a novel RS-related protein that interacts with RNA and RNA binding proteins. Nucleic Acids Res 2014;42:3177-93
  • Giovannone B, Lee E, Laviola L, et al. Two novel proteins that are linked to insulin-like growth factor (IGF-I) receptors by the Grb10 adapter and modulate IGF-I signaling. J Biol Chem 2003;278:31564-73
  • Morita M, Ler LW, Fabian MR, et al. A novel 4EHP-GIGYF2 translational repressor complex is essential for mammalian development. Mol Cell Biol 2012;32:3585-93
  • Lee TH, Wisniewski HG, Vilcek J. A novel secretory tumor necrosis factor-inducible protein (TSG-6) is a member of the family of hyaluronate binding proteins, closely related to the adhesion receptor CD44. J Cell Biol 1992;116:545-57
  • Milner CM, Higman VA, Day AJ. TSG-6: a pluripotent inflammatory mediator? Biochem Soc Trans 2006;34(Pt 3):446-50
  • Lang F, Voelkl J. Therapeutic potential of serum and glucocorticoid inducible kinase inhibition. Expert Opin Investig Drugs 2013;22:701-14
  • Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401-4
  • Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1

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.