Next Article in Journal
ABA Mediates Plant Development and Abiotic Stress via Alternative Splicing
Next Article in Special Issue
The Interplay between PARP Inhibitors and Immunotherapy in Ovarian Cancer: The Rationale behind a New Combination Therapy
Previous Article in Journal
Overcoming Steroid Resistance in Pediatric Acute Lymphoblastic Leukemia—The State-of-the-Art Knowledge and Future Prospects
Previous Article in Special Issue
Human Papillomaviruses as Infectious Agents in Gynecological Cancers. Oncogenic Properties of Viral Proteins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rare Subtype of Endometrial Cancer: Undifferentiated/Dedifferentiated Endometrial Carcinoma, from Genetic Aspects to Clinical Practice

1
Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch and Chang Gung University College of Medicine, Taoyuan 333, Taiwan
2
Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
3
Department of Pathology, Chang Gung Memorial Hospital, Linkou Branch and Chang Gung University College of Medicine, Taoyuan 333, Taiwan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(7), 3794; https://doi.org/10.3390/ijms23073794
Submission received: 28 February 2022 / Revised: 24 March 2022 / Accepted: 28 March 2022 / Published: 30 March 2022
(This article belongs to the Special Issue New Insights into the Pathophysiology of Gynecological Cancers)

Abstract

:
Endometrial cancer (EC) is one of the most common gynecologic cancers worldwide. There were 417,367 newly diagnosed cases and 97,370 deaths due to this disease worldwide in 2020. The incidence rates have increased over time, especially in countries with rapid socioeconomic transitions, and EC has been the most prevalent gynecologic malignancy in Taiwan since 2012. The new EC molecular classifications of The Cancer Genome Atlas (TCGA) Research Network include clear-cell carcinoma, serous carcinoma, and carcinosarcoma, while undifferentiated/dedifferentiated EC (UDEC) is not mentioned, and most previous clinical trials for EC have not included UDEC. UDEC is rare, has an aggressive growth pattern, tends to be diagnosed at an advanced stage, and is resistant to conventional chemotherapy. In this review, case series or case reports on the clinical features and genomic/epigenetic and expression profiles on UDEC data are summarized in order to identify potential molecular targets for current and future research.

1. Introduction

Endometrial cancer (EC) is one of the most common gynecologic cancers worldwide. There were 61,880 newly diagnosed EC cases and 12,160 related deaths in the United States in 2019 [1], and EC has been the most prevalent gynecologic malignancy in Taiwan since 2012 [2]. The histologic subtype is prognostic; however, most previous studies on this topic have considered the high-risk subtypes to be serous, clear-cell, and carcinosarcoma. The Cancer Genome Atlas (TCGA) Research Network has provided new molecular classifications of EC [3], with profound impacts for clinical practice, but undifferentiated/dedifferentiated EC (UDEC) was not mentioned. Silva et al. reported that low-grade endometrioid carcinoma (grade 1 and 2) comprised 71% of EC, high-grade (HG), and non-endometrioid types in the remaining 29% [4]. Among the remaining 29% of the non-endometrioid type, the incidence was 13% serous carcinoma, 7% clear-cell carcinoma, and 9% undifferentiated EC (UEC) in the MD Anderson series [4]. The UEC is a solid-pattern tumor without specific morphologic evidence of epithelial differentiation [5]. It has an aggressive growth pattern that tends to be diagnosed at an advanced International Federation of Gynecology and Obstetrics (FIGO) stage and is resistant to conventional chemotherapy [4]. Dedifferentiated EC (DEC) is characterized by the coexistence of low-grade EC and UEC [6]. DEC has not been widely recognized due to its solid part usually being misdiagnosed as a grade 3 endometrioid EC [6]. A worse outcome than grade 3 EC was also found [6]. In a recent population-based study using the National Cancer Database of the United States (2004–2013), 1.1% of all ECs met the criteria of UDEC, which may reflect an underdiagnosis in earlier years [7].
UDEC harbors specific genetic features different from endometrioid carcinoma. Generally, endometrioid carcinoma is a hormone-dependent tumor that expresses hormone receptors that may respond to hormone therapy. Unfortunately, UDEC seldom has detectable hormone receptors and its tumorigenesis pathway has distinct features, such as microsatellite instability (MSI-H)/mismatch repair (MMR) protein [8,9] and the genomic inactivation of core components of the SWI/SNF chromatin-remodeling complex [10,11].
Thus, our aim is to review the literature concerning clinical features and molecular targets for novel therapy.

2. Histological Diagnosis, Immunohistochemistry, and Genetic Alterations

2.1. Histological Diagnosis, Immunohistochemistry

Histologically, UEC is characterized by a proliferation of dyscohesive tumor cells growing in patternless, sheet-like arrangements [12,13]. The tumor cells are typically medium-sized and monomorphic [12,13]. However, occasionally, marked nuclear pleomorphism, vague spindling, foci of abrupt keratinization, rhabdoid cells with abundant eosinophilic cytoplasm and eccentric nuclei, or a myxoid stroma can be noted [12,13]. A differentiated component, most commonly FIGO grade 1 or 2 endometrioid carcinoma, can be identified in approximately 40% of UEC cases and is termed “dedifferentiated carcinoma” [5]. The tumor histology poses a great diagnostic challenge and misdiagnosis is not uncommon, as it can mimic grade 3 EC, neuroendocrine carcinoma, carcinosarcoma, endometrial stromal sarcoma, undifferentiated sarcoma, melanoma, lymphoma, and plasmacytoma [4,5,13]. Immunohistochemically, UEC typically shows the focal expression of epithelial markers, especially CK18 and epithelial membrane antigen (EMA), the loss of cell adhesion molecules such as E-cadherin and claudin-4, and a lack of expression of Müllerian markers such as PAX8, ER, and PR [11,12,13,14]. A panel including PAX8, E-cadherin, cytokeratin, and EMA can be helpful to distinguish between UEC and grade 3 EC [5]. DNA MMR protein (MLH1, MSH2, MSH6, and PMS2) expression is lost in 44% of UEC cases [15]. Approximately 60% of UEC cases lose the expression of switch/sucrose non-fermenting (SWI/SNF) complex proteins, including ARID1B, SMARCA4 (BRG1), and SMARCB1 (INI1) [16,17]. Recent immunochemistry stain findings of the above markers are listed in Table 1. SWI/SNF complex deficiency ranged from 20.0 to 69.4%, and MMR deficiency (MMRd) ranged from 46.2 to 73.3% across different studies [8,11,18,19,20,21].

2.2. TCGA Molecular Classification in UDEC

TCGA documented four molecular categories of EC, known as POLE/ultramutated, microsatellite instability (MSI)/hypermutated, copy-number-low/TP53-wild-type, and copy-number-high/p53-abnormal [3]. A systemic review using pooled case series data reclassified into TCGA subgroups of UDEC (n = 73) found 44.0% of cases with MMR deficiency (MMR-d), 12.4% with POLE mutation, and 18.6% with p53 abnormality [22], for which the proportion of UDEC in the MSI group was lower than the previous report (about 60%) [23,24] under this calculation but was consistently higher than the overall EC (28%) in the TCGA cohort [3]. The proportion of UDEC in the POLE group (12.4%) was also higher than EC (7.3%) in the TCGA cohort without UDEC. As the majority of UDECs belonged to the MSI and POLE groups, a high mutational load is a feature of UDECs different from other high-risk histological subtypes of EC [22]. Since a high mutational load is a predictor of response to immunotherapy, UDECs may benefit from such treatment [8,25].

2.3. Switch/Sucrose Non-Fermentable (SWI/SNF) Complex Proteins

As component proteins of the switch/sucrose non-fermenting (SWI/SNF) chromatin remodeling complex, BRG1 (encoded by SMARCA4) and INI1 (encoded by SMARCB1) mutations are known to be related to several types of human malignancies [26,27]. They are key regulators of nucleosome positioning [28]. SWI/SNF complexes generate energy from ATP hydrolysis to slide or eject nucleosomes [29]. Inactivating mutations involving SMARCA4 or SMARCB1 cause the loss of BRG1 or INI1 expression, which is a significant subset of DEC [10,30]. The loss of BRG1 or INI1 is also associated with the loss of PAX8 and estrogen receptor (ER) in the undifferentiated component [23]. Mutations in the genes encoding SWI/SNF subunits often result in a loss of function. Missense mutations might be the most common type of mutation, for example, within the enzymatic ATPase domain of SMARCA4 [31]. The SMARCA4-deficient and SMARCB1-deficient DECs were reported to have a prevalence of approximately 50–68% [11,30]. Accompanied with these mutations, the majority (66–73%) of the undifferentiated component developed in a mismatch repair-deficient molecular context [11,30]. The SMARCA4-deficient and SMARCB1-deficient types were also associated with a worse outcome [11].

2.4. ARID1A and ARID1B

ARID1A and ARID1B are alternate but obligatory DNA-binding subunits of the SWI/SNF complex. Concurrent inactivating mutations that result in the loss of both ARID1A and ARID1B are expected to block chromatin remodeling functions, resulting in difficulty in binding and targeting DNA [23]. ARID1A and ARID1B co-inactivation appears to be an alternate mechanism to BRG1 or INI1, which results in the occurrence of the undifferentiated component [23]. Overall, ARID1A is the most frequently mutated SWI/SNF subunit across cancer types [29] and has a tumor-suppressive function, whereby it triggers cancer development by interfering with the DNA-damage response and cell cycle pathways [32,33]. Furthermore, several studies have also demonstrated that an ARID1A loss is associated with the activation of phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) and the concurrent loss of PTEN expression, which both activate the PI3K/AKT/ mTOR cell cycle pathway [34,35,36,37]. A greater propensity for MMRd EC was observed in 75% of these ARID1A/ARID1B co-deficient DECs [23].

2.5. Deficiency of MMR Protein

Known MMR proteins include mutL protein homolog 1 (MLH1), postmeiotic segregation increased 2 (PMS2), mutS protein homolog 2 (MSH2), and mutS protein homolog 6 (MSH6). MMR defects were significantly associated with poor clinical outcomes in all types of EC. A higher tumor grade and the presence of lymphovascular space invasion (LVSI) were related to both epigenetic MMR defects and mutations [38]. However, MMRd was also related to better adjuvant treatment response and may have favorable survival [39]. Tafe et al. reviewed IHC in 17 cases of UEC from endometrium and ovary and showed that eight cases (47%) had MMRd UEC [13]. In a pooled cohort of UDEC (n = 73), 44% of cases were in the MMRd group [22].

2.6. Programmed Cell Death Ligand-1 (PD-L1)

Programmed death receptor (PD-1) and its ligand PD-L1 are known as important biomarkers for immune checkpoint inhibitors. As a co-inhibitory transmembrane receptor expressed by T cells, PD-1 can inhibit the proliferation, survival, and cytokine production of T cells by engaging with PD-L1 [40], resulting in immune escape. Extensive work has been undertaken to investigate PD-1 blockage in a variety of cancers, leading to clinical approval for treatment, for example, melanoma with pembrolizumab, and renal cell carcinoma with nivolumab [41]. However, in 2021, a large cohort with 833 samples reported that 45% positivity of PD-L1 (combined positive score, CPS > 1) may relate to a better outcome in high-risk EC (including seven UDECs) [42]. Some studies found a positive correlation with MMR-d- and PD-L1-positive tumors. Sloan et al. also found 100% PD-L1 positivity in MMRd patients, and 66% PD-L1 positivity in MMRp patients [43]. In the undifferentiated component, nine cases (53%) belonged to the MMRd group and all of these cases exhibited PD-L1 expression. Not only in all ECs, but also in UDEC, MMRd was significantly associated with PD-L1 expression (p = 0.026) [44]. However, in an analysis of 17 patients from a cancer center in Jordan, there was no correlation with MMR-p and PD-L1. However, PD-L1 expression in UDEC in this series accounted for 66.7% of cases (10/17 cases) [18], which suggests that DEC could be a good target for immune checkpoint inhibitors [44]. A DEC case was reported to have been successfully salvaged with pembrolizumab, with 15 months of progression-free survival [45].

2.7. Molecular Genetic Heterogeneity

The UDEC, as previously described, are genetically heterogeneous, with cases belonging to all four groups of TCGA classification. To further investigate genetic alteration, Rosa-Rosa, J.M. et al. carried out exome sequencing in 18 samples of UDEC. MSI hypermutation accounted for 44% (8/18) of UDEC cases, and POLE mutation for 11% (2/18); PTEN mutation accounted for 28% of endometrioid-like ECs (5/18); TP53 mutation accounted for 11% of serous-like ECs (2/18); and there was one unclassified case [46]. The MSI group was larger than most series of sporadic EC (around 30%) [47]. One of the POLE groups in this series (two cases) harbored a TP53 mutation. One-third of UEC in this series were low-copy-number endometrial carcinomas. The most frequent mutation was the PTEN mutation, associated with the PIK3CA pathway. Another report of sequencing analysis results also found UDEC with somatic mutations in PIK3CA (50%), CTNNB1 (30%), TP53 (30%), FBXW7 (20%), and PPP2R1A (20%) [48].

3. Treatment and Prognosis

Due to the rarity of UDEC, there was little experience with novel treatment in case series. Real-world experience was limited to conventional chemotherapy and/or radiotherapy [49,50], as listed in Table 2 [11,21,45,49,50,51]. According to most clinical guidelines [52,53], comprehensive staging surgery should be performed in operable cases, including total hysterectomy with bilateral salpingo-oophorectomy, systematic lymphadenectomy, and infracolic omentectomy [53]. UDEC is classified as a type II EC, and warrants chemotherapy even at an early stage. Paclitaxel and carboplatin are suggested for stage IA with myometrial invasion every three weeks for 3–6 cycles. In patients with LVSI, brachytherapy may be arranged. For stage IB to IIIC2, pelvic radiotherapy with brachytherapy followed by chemotherapy is recommended. For positive pelvic or para-aortic lymph nodes, extended field radiotherapy should be arranged [19].
Despite such a heavy treatment setting, a fulminant recurrence pattern with short survival was reported by Han et al. All four patients in this series were LVSI positive. One stage II and one stage IIIB patient relapsed and died within a short interval. Although one patient (stage II) refused adjuvant therapy, the other patient with stage IIIB and vaginal wall extension received complete chemotherapy and radiotherapy, but still relapsed one month after she had completed treatment [50].
Pfaendler treated two chemoresistant UEC patients with a rapid progression pattern. As no hormone receptor was present, nor a microsatellite stable feature, case 1 with stage IA, LVSI-negative UEC, and 20% myometrial invasion, received six cycles of paclitaxel and carboplatin, but still had recurrence after 6 months, and was deceased 6 weeks after recurrence. In case 2, with MLH1 and PMS2 loss, pembrolizumab was tried for one dose, but the patient moved overseas [49].
Most cases of DEC coexisted with UEC of low-grade (FIGO grade 1-2) EC (DEC-LG). However, DEC can arise with a background of high-grade carcinoma (DEC-HG). A case series (n = 18) compared DEC-HG with DEC-LG, in which DEC-HG presented with advanced disease (stage III–IV) (7/11, 64%), whereas most cases of DEC-LG were stage 1 (6/7, 86%). A total of four DEC-HG patients recurred or died, while only one DEC-LG patient recurred, with a mean follow-up of 23.2 months [21]. Goh et al. reported seven DEC cases in Singapore, four of which had recurrent/progressive disease. A case with FIGO stage IIIA DEC had extensive recurrence affecting the thorax, abdomen, and pelvis after surgery and chemotherapy. She received pembrolizumab and gemcitabine and survived for a further 10 months under stable disease and the resolution of ascites. The other three cases of recurrent/progressive disease all died, even with salvage efforts. The only long-term survivor with a favorable outcome of an OS of 56 months was a stage IIIC1 patient. Her tumor size was relatively small (1.8cm), with a 25% UEC component and one (out of eight) lymph node metastasis of the endometrioid component. They also compared DEC patients with grade 3 endometrioid EC, which showed a 2-year survival OS of 31.3% in DEC versus 82.8% in their institution [45]. Whether the percentage of DEC and the grade of the DEC component in the primary tumor affects prognosis is still controversial.
The genomic inactivation of core SWI/SNF complex protein was recently shown to account for two-thirds of DEC/UECs. When comparing SWI/SNF-deficient tumors with SWI/SNF-intact tumors, a poorer prognosis in the SWI/SNF-deficient group was noted. The 2-year disease-specific survival (DSS) for stages I and II disease was 65 % in deficient tumors and 100% in intact tumors (p = 0.042). For stages III and IV disease, the medial survival was 4 months for SWI/SNF-deficient tumors (36 months for intact tumors, p = 0.0003). SWI/SNF-deficient tumors possessed a highly progressive disease pattern and were resistant to conventional chemotherapy [11]. Further targets or immune therapies should be attempted, aside from chemotherapy.
In a recent case series of 52 UDECs, in which MMR status was checked, 30 of 43 (69.8%) were MMRd. The 5-year disease-free survival was 80% for stage I/II, 29% for stage III, and 10% for stage IV. Multivariate analysis suggested that adjuvant chemotherapy, adjuvant radiotherapy, and a lower FIGO stage contributed to better disease-free survival (p < 0.05) [19].

4. Preclinical Data and Relevant Clinical Data

Several new drugs have been tested in UDEC in preclinical studies. Fibroblast Growth Factor Receptor (FGFR2) mutation was identified in approximately 10% of patients with primary endometrial cancer and more than 90% of patients with PIK3CA activation [54]. Thus, they tried a pan-FGFR inhibitor (BGJ398, infigratinib) with pan-PI3K inhibitors (GDC-0941, BKM 120) in endometrial cancer cell lines, which showed a significant increase in cell death and long-term survival when the PI3K inhibitor was combined with BGJ398 [54]. Lin et al. performed whole-exome sequencing from specimens of a uterine DEC patient, which revealed an FGFR2 mutation and CCNE2 amplification. The former was targeted with the FGFR inhibitor lenvatinib, while the latter was treated with the cell cycle inhibitor palbociclib in patient-derived xenograft (PDX) mice with UDEC. Compared with control mice, the tumor shrank significantly in the treatment arm [55]. The first approved FGFR2 inhibitor, pemigatinib, for treating cholangiocarcinoma [56] is now used in recurring FGFR-mutated solid tumors, including EC (NCT02393248, FIGHT-101). For a rare disease such as UDEC, the use of a precise genetic survey can find an effective target therapy for a specific molecular alteration of cancer.
Targeting the SWI/SNF complex may be the next step of treating UDEC, since two-thirds of UDEC patients had SWI/SNF deficiency [11,30]. Several molecules that inhibit SWI/SNF ATPase activity have been reported. The first published inhibitor was PFI-3 [57], but due to the lack of chemical stability in cellular systems over longer time periods, several other SMARCA2/4 BRD inhibitors are currently under development [58,59]. Proteolysis-targeting chimeras induce ubiquitin transfer onto target proteins, thereby marking them for proteasomal degradation. This was applied to target BRD9, a SWI/SNF complex subunit [33]. For example, AU-15330 induced a potent inhibitor of tumor growth in xenograft models of prostate cancer, and synergized with the androgen receptor antagonist enzalutamide, even inducing disease remission in castration-resistant prostate cancer models without toxicity [60].
Aurora A was reported to be a potential therapeutic target in ARID1A-deficient colorectal cancer cells [61]. Wild-type ARID1A downregulated the expression of Aurora A, while ARID1A loss led to increased expression levels of Aurora A and activated the cell division cycle. Thus, the inhibition of Aurora A with a kinase inhibitor induced a G2/M phase arrest, followed by apoptosis [33]. Alisertib as an Aurora A kinase inhibitor has recently been used in phase 3 randomized trials for hematologic malignancy. While it showed disappointing early results, it has been investigated in a number of cancer types, as a monotherapy or in combination [62]. In a randomized phase 3 trial, alpelisib plus fulvestrant were tested in PIK3CA-mutated, hormone-receptor-positive advanced breast cancer, which prolonged progression-free survival (11.0 months vs. 5.7 months) for longer than in the placebo plus fulvestrant arm [63].
The enhancer of zeste homologue 2 (EZH2) inhibitor tazemetostat is under testing in adults with diffuse large B cell lymphoma or SMARCB1-negative or SMARCA4-negative solid tumors, and in pediatric patients with rhabdoid tumors, synovial sarcoma, epithelioid sarcoma, or other cancers with SMARCB1, SMARCA4, or EZH2 mutations [64,65,66,67]. A trial was performed in patients with epithelioid sarcomas (nearly all tumors have homozygous deletion of SMARCB1), which demonstrated a response rate of 15%, and of the patients who responded, 67% had a response lasting 6 months or longer [68]. However, applications in EC with the SWI/SNF mutation remained unknown, until now.
Finally, as we know, MMR-d and PD-L1 account for about 60% of these UDECs. Immune checkpoint inhibitors should be considered, although there is a lack of trials or large databases grounded in real world experience specific for UEC/DEC. In recent EC trials, KENOTE-158 used pembrolizumab monotherapy for noncolorectal high-microsatellite-instability/MMR-d cancers, which included 49 cases (21.0%) of endometrial cancer and yielded the best response rate (57.1%) and longest progression-free survival (25.7 months) among gastric, cholangiocarcinoma, pancreatic, small intestine, ovarian, and brain cancer [69]. Pembrolizumab plus levatinib showed a promising antitumor effect in KEYNOTE-146, which included one UDEC patient [70], and has further been verified by a phase 3 trial, KEYNOTE-775, presented at the 2021 SGO conference [71]. Another anti-PD-1 antibody, dostarlimab, showed a 42% objective response rate in MMRd EC in its phase 1 trial [72]. APR-246, a prodrug that binds to cysteine residues in mutant p53 and restores its wild-type function, showed synergistic effects with chemotherapy in ovarian cancer cell lines and resensitized platinum-resistant ovarian cancer cells [73]. Adavosertib (MK-1775), a potent, small-molecule WEE1 kinase inhibitor, showed an antitumor effect in combination with chemotherapy and radiotherapy in preclinical studies. There are several ongoing trials of APR-246 and adavosertib in ovarian cancer [73]. Since a TP53 mutation is one of the features of UEDC [22,48], agents targeting the mutant p53 pathway can also be explored.
We summarize the potential therapeutic agents targeting members of these pathways in Figure 1. Target/immune therapies are collected and depicted in Table 3.

5. Conclusions and Future Directions

UDECs are clinically aggressive diseases with chemoresistance. Recurrent disease almost always has a dismal outcome. The prevalence of UDEC might have been underestimated, and the prevalence of DEC-HG versus DEC-LG needs more research for clarification. UDEC patients should be encouraged to enroll in EC clinical trials, and the sponsors should be able to conduct subgroup analyses for UDEC. Since about half of these tumors have MMR-d features and the expression of PD-L1, immune therapy may be eligible for treatment options. The SWI/SNF complex plays a crucial role, especially in patients with a poor prognosis. Targeting the SWI/SNF pathway and subunit may open a new therapeutic window for UDECs. Comprehensive genetic sequencing is also helpful in finding possible potent drugs.

6. Materials and Methods

We searched articles published up to February 2022 in the PubMed (https://pubmed.ncbi.nlm.nih.gov), UpToDate, and Google Scholar databases by using the following keywords: undifferentiated or dedifferentiated carcinoma of uterus, genetic analysis, molecular markers, and prognosis and treatment of UDEC. The most relevant reviews and case series were included, and the reference lists of these selected articles were read. By focusing on future directions, the latest case reports or preclinical data of special or target therapies were also presented.

Author Contributions

H.-J.T., R.-C.W., C.-Y.L. and C.-H.L. all contributed to the concept, design, literature review, draft, and final approval of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Chang Gung Foundation (CMRPG3L1021).

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Health Promotion Administration Ministry of Health and Welfare Taiwan. 2018 Cancer Registry Annual Report. Available online: https://www.hpa.gov.tw/Pages/ashx/File.ashx?FilePath=~/File/Attach/6069/File_5962.pdf (accessed on 25 January 2022).
  3. The Cancer Genome Atlas Research Network; Levine, D.A. Integrated genomic characterization of endometrial carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Silva, E.G.; Deavers, M.T.; Malpica, A. Undifferentiated carcinoma of the endometrium: A review. Pathology 2007, 39, 134–138. [Google Scholar] [CrossRef] [PubMed]
  5. Murali, R.; Davidson, B.; Fadare, O.; Carlson, J.A.; Crum, C.P.; Gilks, C.B.; Irving, J.A.; Malpica, A.; Matias-Guiu, X.; McCluggage, W.G.; et al. High-grade Endometrial Carcinomas: Morphologic and Immunohistochemical Features, Diagnostic Challenges and Recommendations. Int. J. Gynecol. Pathol. 2019, 38 (Suppl. 1), S40–S63. [Google Scholar] [CrossRef]
  6. Li, Z.; Zhao, C. Clinicopathologic and Immunohistochemical Characterization of Dedifferentiated Endometrioid Adenocarcinoma. Appl. Immunohistochem. Mol. Morphol. 2016, 24, 562–568. [Google Scholar] [CrossRef]
  7. AlHilli, M.; Elson, P.; Rybicki, L.; Amarnath, S.; Yang, B.; Michener, C.M.; Rose, P.G. Undifferentiated endometrial carcinoma: A National Cancer Database analysis of prognostic factors and treatment outcomes. Int. J. Gynecol. Cancer 2019, 29, 1126–1133. [Google Scholar] [CrossRef]
  8. Hacking, S.; Jin, C.; Komforti, M.; Liang, S.; Nasim, M. MMR deficient undifferentiated/dedifferentiated endometrial carcinomas showing significant programmed death ligand-1 expression (sp 142) with potential therapeutic implications. Pathol. Res. Pract. 2019, 215, 152552. [Google Scholar] [CrossRef]
  9. Kaur, R.; Mehta, J.; Borges, A.M. Role of SMARCA4 (BRG1) and SMARCB1 (INI1) in Dedifferentiated Endometrial Carcinoma With Paradoxical Aberrant Expression of MMR in the Well-Differentiated Component: A Case Report and Review of the Literature. Int. J. Surg. Pathol. 2021, 29, 571–577. [Google Scholar] [CrossRef]
  10. Strehl, J.D.; Wachter, D.L.; Fiedler, J.; Heimerl, E.; Beckmann, M.W.; Hartmann, A.; Agaimy, A. Pattern of SMARCB1 (INI1) and SMARCA4 (BRG1) in poorly differentiated endometrioid adenocarcinoma of the uterus: Analysis of a series with emphasis on a novel SMARCA4-deficient dedifferentiated rhabdoid variant. Ann. Diagn. Pathol. 2015, 19, 198–202. [Google Scholar] [CrossRef]
  11. Tessier-Cloutier, B.; Coatham, M.; Carey, M.; Nelson, G.S.; Hamilton, S.; Lum, A.; Soslow, R.A.; Stewart, C.J.; Postovit, L.M.; Kobel, M.; et al. SWI/SNF-deficiency defines highly aggressive undifferentiated endometrial carcinoma. J. Pathol. Clin. Res. 2021, 7, 144–153. [Google Scholar] [CrossRef]
  12. World Health Organization. Female Genital Tumours: WHO Classification of Tumours, 5th ed.; International Agency for Research on Cancer: Lyon, France; WHO Press: Geneva, Switzerland, 2020; Volume 4. [Google Scholar]
  13. Tafe, L.J.; Garg, K.; Chew, I.; Tornos, C.; Soslow, R.A. Endometrial and ovarian carcinomas with undifferentiated components: Clinically aggressive and frequently underrecognized neoplasms. Mod. Pathol. 2010, 23, 781–789. [Google Scholar] [CrossRef]
  14. Zhang, X.; Yu, M. Undifferentiated endometrial carcinoma: A selected immunohistochemical panel including PAX-8 and E-cadherin for aiding distinction from other endometrial carcinomas. Ann. Diagn. Pathol. 2019, 39, 36–41. [Google Scholar] [CrossRef] [PubMed]
  15. Santoro, A.; Angelico, G.; Travaglino, A.; Inzani, F.; Arciuolo, D.; Valente, M.; D’Alessandris, N.; Scaglione, G.; Fiorentino, V.; Raffone, A.; et al. New Pathological and Clinical Insights in Endometrial Cancer in View of the Updated ESGO/ESTRO/ESP Guidelines. Cancers 2021, 13, 2636. [Google Scholar] [CrossRef] [PubMed]
  16. Kobel, M.; Hoang, L.N.; Tessier-Cloutier, B.; Meng, B.; Soslow, R.A.; Stewart, C.J.R.; Lee, C.H. Undifferentiated Endometrial Carcinomas Show Frequent Loss of Core Switch/Sucrose Nonfermentable Complex Proteins. Am. J. Surg. Pathol. 2018, 42, 76–83. [Google Scholar] [CrossRef] [PubMed]
  17. Kihara, A.; Amano, Y.; Matsubara, D.; Fukushima, N.; Fujiwara, H.; Niki, T. BRG1, INI1, and ARID1B Deficiency in Endometrial Carcinoma: A Clinicopathologic and Immunohistochemical Analysis of a Large Series From a Single Institution. Am. J. Surg. Pathol. 2020, 44, 1712–1724. [Google Scholar] [CrossRef] [PubMed]
  18. Al-Hussaini, M.; Lataifeh, I.; Jaradat, I.; Abdeen, G.; Otay, L.; Badran, O.; Abu Sheikha, A.; Dayyat, A.; El Khaldi, M.; Ashi Al-Loh, S. Undifferentiated Endometrial Carcinoma, an Immunohistochemical Study Including PD-L1 Testing of a Series of Cases From a Single Cancer Center. Int. J. Gynecol. Pathol. 2018, 37, 564–574. [Google Scholar] [CrossRef] [PubMed]
  19. Hamilton, S.N.; Tinker, A.V.; Kwon, J.; Lim, P.; Kong, I.; Sihra, S.; Koebel, M.; Lee, C.H. Treatment and outcomes in undifferentiated and dedifferentiated endometrial carcinoma. J. Gynecol. Oncol. 2022, 33, e25. [Google Scholar] [CrossRef]
  20. Hoang, L.N.; Lee, Y.S.; Karnezis, A.N.; Tessier-Cloutier, B.; Almandani, N.; Coatham, M.; Gilks, C.B.; Soslow, R.A.; Stewart, C.J.; Kobel, M.; et al. Immunophenotypic features of dedifferentiated endometrial carcinoma-insights from BRG1/INI1-deficient tumours. Histopathology 2016, 69, 560–569. [Google Scholar] [CrossRef]
  21. Busca, A.; Parra-Herran, C.; Nofech-Mozes, S.; Djordjevic, B.; Ismiil, N.; Cesari, M.; Nucci, M.R.; Mirkovic, J. Undifferentiated endometrial carcinoma arising in the background of high-grade endometrial carcinoma-Expanding the definition of dedifferentiated endometrial carcinoma. Histopathology 2020, 77, 769–780. [Google Scholar] [CrossRef]
  22. Travaglino, A.; Raffone, A.; Mascolo, M.; Guida, M.; Insabato, L.; Zannoni, G.F.; Zullo, F. TCGA Molecular Subgroups in Endometrial Undifferentiated/Dedifferentiated Carcinoma. Pathol. Oncol. Res. 2020, 26, 1411–1416. [Google Scholar] [CrossRef]
  23. Coatham, M.; Li, X.; Karnezis, A.N.; Hoang, L.N.; Tessier-Cloutier, B.; Meng, B.; Soslow, R.A.; Blake Gilks, C.; Huntsman, D.G.; Stewart, C.J.; et al. Concurrent ARID1A and ARID1B inactivation in endometrial and ovarian dedifferentiated carcinomas. Mod. Pathol. 2016, 29, 1586–1593. [Google Scholar] [CrossRef] [PubMed]
  24. Stewart, C.J.; Crook, M.L. SWI/SNF complex deficiency and mismatch repair protein expression in undifferentiated and dedifferentiated endometrial carcinoma. Pathology 2015, 47, 439–445. [Google Scholar] [CrossRef] [PubMed]
  25. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Masliah-Planchon, J.; Bieche, I.; Guinebretiere, J.M.; Bourdeaut, F.; Delattre, O. SWI/SNF chromatin remodeling and human malignancies. Annu. Rev. Pathol. 2015, 10, 145–171. [Google Scholar] [CrossRef] [PubMed]
  27. Shain, A.H.; Pollack, J.R. The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS ONE 2013, 8, e55119. [Google Scholar] [CrossRef] [Green Version]
  28. Euskirchen, G.; Auerbach, R.K.; Snyder, M. SWI/SNF chromatin-remodeling factors: Multiscale analyses and diverse functions. J. Biol. Chem. 2012, 287, 30897–30905. [Google Scholar] [CrossRef] [Green Version]
  29. Mittal, P.; Roberts, C.W.M. The SWI/SNF complex in cancer-biology, biomarkers and therapy. Nat. Rev. Clin. Oncol. 2020, 17, 435–448. [Google Scholar] [CrossRef]
  30. Karnezis, A.N.; Hoang, L.N.; Coatham, M.; Ravn, S.; Almadani, N.; Tessier-Cloutier, B.; Irving, J.; Meng, B.; Li, X.; Chow, C.; et al. Loss of switch/sucrose non-fermenting complex protein expression is associated with dedifferentiation in endometrial carcinomas. Mod. Pathol. 2016, 29, 302–314. [Google Scholar] [CrossRef]
  31. Hodges, H.C.; Stanton, B.Z.; Cermakova, K.; Chang, C.Y.; Miller, E.L.; Kirkland, J.G.; Ku, W.L.; Veverka, V.; Zhao, K.; Crabtree, G.R. Dominant-negative SMARCA4 mutants alter the accessibility landscape of tissue-unrestricted enhancers. Nat. Struct. Mol. Biol. 2018, 25, 61–72. [Google Scholar] [CrossRef]
  32. Pavlidou, E.N.; Balis, V. Diagnostic significance and prognostic role of the ARID1A gene in cancer outcomes (Review). World Acad. Sci. J. 2020, 2, 49–64. [Google Scholar] [CrossRef] [Green Version]
  33. Wanior, M.; Krämer, A.; Knapp, S.; Joerger, A.C. Exploiting vulnerabilities of SWI/SNF chromatin remodelling complexes for cancer therapy. Oncogene 2021, 40, 3637–3654. [Google Scholar] [CrossRef] [PubMed]
  34. Samartzis, E.P.; Gutsche, K.; Dedes, K.J.; Fink, D.; Stucki, M.; Imesch, P. Loss of ARID1A expression sensitizes cancer cells to PI3K- and AKT-inhibition. Oncotarget 2014, 5, 5295–5303. [Google Scholar] [CrossRef] [PubMed]
  35. Lee, D.; Yu, E.J.; Ham, I.H.; Hur, H.; Kim, Y.S. AKT inhibition is an effective treatment strategy in ARID1A-deficient gastric cancer cells. Onco Targets Ther. 2017, 10, 4153–4159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Chandler, R.L.; Damrauer, J.S.; Raab, J.R.; Schisler, J.C.; Wilkerson, M.D.; Didion, J.P.; Starmer, J.; Serber, D.; Yee, D.; Xiong, J.; et al. Coexistent ARID1A-PIK3CA mutations promote ovarian clear-cell tumorigenesis through pro-tumorigenic inflammatory cytokine signalling. Nat. Commun. 2015, 6, 6118. [Google Scholar] [CrossRef] [Green Version]
  37. Bosse, T.; ter Haar, N.T.; Seeber, L.M.; van Diest, P.J.; Hes, F.J.; Vasen, H.F.; Nout, R.A.; Creutzberg, C.L.; Morreau, H.; Smit, V.T. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations, TP53 and microsatellite instability in endometrial cancer. Mod. Pathol. 2013, 26, 1525–1535. [Google Scholar] [CrossRef] [Green Version]
  38. McMeekin, D.S.; Tritchler, D.L.; Cohn, D.E.; Mutch, D.G.; Lankes, H.A.; Geller, M.A.; Powell, M.A.; Backes, F.J.; Landrum, L.M.; Zaino, R.; et al. Clinicopathologic Significance of Mismatch Repair Defects in Endometrial Cancer: An NRG Oncology/Gynecologic Oncology Group Study. J. Clin. Oncol. 2016, 34, 3062–3068. [Google Scholar] [CrossRef]
  39. Shikama, A.; Minaguchi, T.; Matsumoto, K.; Akiyama-Abe, A.; Nakamura, Y.; Michikami, H.; Nakao, S.; Sakurai, M.; Ochi, H.; Onuki, M.; et al. Clinicopathologic implications of DNA mismatch repair status in endometrial carcinomas. Gynecol. Oncol. 2016, 140, 226–233. [Google Scholar] [CrossRef]
  40. Amarin, J.Z.; Mansour, R.; Al-Ghnimat, S.; Al-Hussaini, M. Differential Characteristics and Prognosis of PD-L1-Positive Endometrial Carcinomas: A Retrospective Chart Review. Life 2021, 11, 1047. [Google Scholar] [CrossRef]
  41. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [Green Version]
  42. Zong, L.; Sun, Z.; Mo, S.; Lu, Z.; Yu, S.; Xiang, Y.; Chen, J. PD-L1 expression in tumor cells is associated with a favorable prognosis in patients with high-risk endometrial cancer. Gynecol. Oncol. 2021, 162, 631–637. [Google Scholar] [CrossRef]
  43. Sloan, E.A.; Ring, K.L.; Willis, B.C.; Modesitt, S.C.; Mills, A.M. PD-L1 Expression in Mismatch Repair-deficient Endometrial Carcinomas, Including Lynch Syndrome-associated and MLH1 Promoter Hypermethylated Tumors. Am. J. Surg. Pathol. 2017, 41, 326–333. [Google Scholar] [CrossRef] [PubMed]
  44. Ono, R.; Nakayama, K.; Nakamura, K.; Yamashita, H.; Ishibashi, T.; Ishikawa, M.; Minamoto, T.; Razia, S.; Ishikawa, N.; Otsuki, Y.; et al. Dedifferentiated Endometrial Carcinoma Could be A Target for Immune Checkpoint Inhibitors (Anti PD-1/PD-L1 Antibodies). Int. J. Mol. Sci. 2019, 20, 3744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Goh, C.; Farah, B.L.; Ho, W.Y.; Wong, S.L.; Goh, C.H.R.; Chew, S.H.; Nadarajah, R.; Lim, Y.K.; Ho, T.H. Dedifferentiated endometrioid adenocarcinoma of the uterus: A case series and review of literature. Gynecol. Oncol. Rep. 2020, 32, 100538. [Google Scholar] [CrossRef] [PubMed]
  46. Rosa-Rosa, J.M.; Leskela, S.; Cristobal-Lana, E.; Santon, A.; Lopez-Garcia, M.A.; Munoz, G.; Perez-Mies, B.; Biscuola, M.; Prat, J.; Esther, O.; et al. Molecular genetic heterogeneity in undifferentiated endometrial carcinomas. Mod. Pathol. 2016, 29, 1390–1398. [Google Scholar] [CrossRef]
  47. Bell, D.W.; Ellenson, L.H. Molecular Genetics of Endometrial Carcinoma. Annu. Rev. Pathol. 2019, 14, 339–367. [Google Scholar] [CrossRef]
  48. Kuhn, E.; Ayhan, A.; Bahadirli-Talbott, A.; Zhao, C.; Shih Ie, M. Molecular characterization of undifferentiated carcinoma associated with endometrioid carcinoma. Am. J. Surg. Pathol. 2014, 38, 660–665. [Google Scholar] [CrossRef]
  49. Pfaendler, K.S.; Randall, L.M. Rapid progression of disease in two cases of undifferentiated endometrial carcinoma. Gynecol. Oncol. Rep. 2019, 27, 65–68. [Google Scholar] [CrossRef]
  50. Han, J.; Ki, E.Y.; Rha, S.E.; Hur, S.; Lee, A. Dedifferentiated endometrioid carcinoma of the uterus: Report of four cases and review of literature. World J. Surg. Oncol. 2017, 15, 17. [Google Scholar] [CrossRef] [Green Version]
  51. Silva, E.G.; Deavers, M.T.; Bodurka, D.C.; Malpica, A. Association of low-grade endometrioid carcinoma of the uterus and ovary with undifferentiated carcinoma: A new type of dedifferentiated carcinoma? Int. J. Gynecol. Pathol. 2006, 25, 52–58. [Google Scholar] [CrossRef]
  52. National Comprehensive Cancer Network. Uterine Neoplasm (Version 1.2022). Available online: https://www.nccn.org/professionals/physician_gls/pdf/uterine.pdf (accessed on 28 February 2022).
  53. Concin, N.; Matias-Guiu, X.; Vergote, I.; Cibula, D.; Mirza, M.R.; Marnitz, S.; Ledermann, J.; Bosse, T.; Chargari, C.; Fagotti, A.; et al. ESGO/ESTRO/ESP guidelines for the management of patients with endometrial carcinoma. Int. J. Gynecol. Cancer 2021, 31, 12–39. [Google Scholar] [CrossRef]
  54. Packer, L.M.; Geng, X.; Bonazzi, V.F.; Ju, R.J.; Mahon, C.E.; Cummings, M.C.; Stephenson, S.A.; Pollock, P.M. PI3K Inhibitors Synergize with FGFR Inhibitors to Enhance Antitumor Responses in FGFR2(mutant) Endometrial Cancers. Mol. Cancer Ther. 2017, 16, 637–648. [Google Scholar] [CrossRef] [Green Version]
  55. Lin, C.Y.; Wu, R.C.; Huang, C.Y.; Lai, C.H.; Chao, A.S.; Li, H.P.; Tsai, C.L.; Kuek, E.J.; Hsu, C.L.; Chao, A. A Patient-Derived Xenograft Model of Dedifferentiated Endometrial Carcinoma: A Proof-of-Concept Study for the Identification of New Molecularly Informed Treatment Approaches. Cancers 2021, 13, 5962. [Google Scholar] [CrossRef] [PubMed]
  56. Rizzo, A.; Ricci, A.D.; Brandi, G. Pemigatinib: Hot topics behind the first approval of a targeted therapy in cholangiocarcinoma. Cancer Treat. Res. Commun. 2021, 27, 100337. [Google Scholar] [CrossRef] [PubMed]
  57. Gerstenberger, B.S.; Trzupek, J.D.; Tallant, C.; Fedorov, O.; Filippakopoulos, P.; Brennan, P.E.; Fedele, V.; Martin, S.; Picaud, S.; Rogers, C.; et al. Identification of a Chemical Probe for Family VIII Bromodomains through Optimization of a Fragment Hit. J. Med. Chem. 2016, 59, 4800–4811. [Google Scholar] [CrossRef] [Green Version]
  58. Wanior, M.; Preuss, F.; Ni, X.; Krämer, A.; Mathea, S.; Göbel, T.; Heidenreich, D.; Simonyi, S.; Kahnt, A.S.; Joerger, A.C.; et al. Pan-SMARCA/PB1 Bromodomain Inhibitors and Their Role in Regulating Adipogenesis. J. Med. Chem. 2020, 63, 14680–14699. [Google Scholar] [CrossRef]
  59. Lu, T.; Hu, J.C.; Lu, W.C.; Han, J.; Ding, H.; Jiang, H.; Zhang, Y.Y.; Yue, L.Y.; Chen, S.J.; Jiang, H.L.; et al. Identification of small molecule inhibitors targeting the SMARCA2 bromodomain from a high-throughput screening assay. Acta Pharmacol. Sin. 2018, 39, 1544–1552. [Google Scholar] [CrossRef] [Green Version]
  60. Xiao, L.; Parolia, A.; Qiao, Y.; Bawa, P.; Eyunni, S.; Mannan, R.; Carson, S.E.; Chang, Y.; Wang, X.; Zhang, Y.; et al. Targeting SWI/SNF ATPases in enhancer-addicted prostate cancer. Nature 2022, 601, 434–439. [Google Scholar] [CrossRef] [PubMed]
  61. Wu, C.; Lyu, J.; Yang, E.J.; Liu, Y.; Zhang, B.; Shim, J.S. Targeting AURKA-CDC25C axis to induce synthetic lethality in ARID1A-deficient colorectal cancer cells. Nat. Commun. 2018, 9, 3212. [Google Scholar] [CrossRef]
  62. Liewer, S.; Huddleston, A. Alisertib: A review of pharmacokinetics, efficacy and toxicity in patients with hematologic malignancies and solid tumors. Expert Opin. Investig. Drugs 2018, 27, 105–112. [Google Scholar] [CrossRef]
  63. André, F.; Ciruelos, E.; Rubovszky, G.; Campone, M.; Loibl, S.; Rugo, H.S.; Iwata, H.; Conte, P.; Mayer, I.A.; Kaufman, B.; et al. Alpelisib for PIK3CA-Mutated, Hormone Receptor-Positive Advanced Breast Cancer. N. Engl. J. Med. 2019, 380, 1929–1940. [Google Scholar] [CrossRef]
  64. Kawano, S.; Grassian, A.R.; Tsuda, M.; Knutson, S.K.; Warholic, N.M.; Kuznetsov, G.; Xu, S.; Xiao, Y.; Pollock, R.M.; Smith, J.S.; et al. Preclinical Evidence of Anti-Tumor Activity Induced by EZH2 Inhibition in Human Models of Synovial Sarcoma. PLoS ONE 2016, 11, e0158888. [Google Scholar] [CrossRef] [PubMed]
  65. Italiano, A.; Soria, J.C.; Toulmonde, M.; Michot, J.M.; Lucchesi, C.; Varga, A.; Coindre, J.M.; Blakemore, S.J.; Clawson, A.; Suttle, B.; et al. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: A first-in-human, open-label, phase 1 study. Lancet Oncol. 2018, 19, 649–659. [Google Scholar] [CrossRef]
  66. Knutson, S.K.; Wigle, T.J.; Warholic, N.M.; Sneeringer, C.J.; Allain, C.J.; Klaus, C.R.; Sacks, J.D.; Raimondi, A.; Majer, C.R.; Song, J.; et al. A selective inhibitor of EZH2 blocks H3K27 methylation and kills mutant lymphoma cells. Nat. Chem. Biol. 2012, 8, 890–896. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, K.H.; Kim, W.; Howard, T.P.; Vazquez, F.; Tsherniak, A.; Wu, J.N.; Wang, W.; Haswell, J.R.; Walensky, L.D.; Hahn, W.C.; et al. SWI/SNF-mutant cancers depend on catalytic and non-catalytic activity of EZH2. Nat. Med. 2015, 21, 1491–1496. [Google Scholar] [CrossRef] [Green Version]
  68. Gounder, M.; Schöffski, P.; Jones, R.L.; Agulnik, M.; Cote, G.M.; Villalobos, V.M.; Attia, S.; Chugh, R.; Chen, T.W.; Jahan, T.; et al. Tazemetostat in advanced epithelioid sarcoma with loss of INI1/SMARCB1: An international, open-label, phase 2 basket study. Lancet Oncol. 2020, 21, 1423–1432. [Google Scholar] [CrossRef]
  69. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
  70. Makker, V.; Taylor, M.H.; Aghajanian, C.; Oaknin, A.; Mier, J.; Cohn, A.L.; Romeo, M.; Bratos, R.; Brose, M.S.; DiSimone, C.; et al. Lenvatinib Plus Pembrolizumab in Patients With Advanced Endometrial Cancer. J. Clin. Oncol. 2020, 38, 2981–2992. [Google Scholar] [CrossRef]
  71. Makker, V. A Multicenter, Open-Label, Randomized, Phase III Study to Compare the Efficacy and Safety of Lenvatinib in Combination with Pembrolizumab Versus Treatment of Physician’s Choice in Patients with Advanced Endometrial Cancer. Available online: https://157slyoyo4y17zpa538hczs1-wpengine.netdna-ssl.com/wp-content/uploads/2021/01/FRIDAY_2021-Virtual-Annual-Meeting-on-Womens-Cancer.pdf (accessed on 28 February 2022).
  72. Oaknin, A.; Tinker, A.V.; Gilbert, L.; Samouëlian, V.; Mathews, C.; Brown, J.; Barretina-Ginesta, M.P.; Moreno, V.; Gravina, A.; Abdeddaim, C.; et al. Clinical Activity and Safety of the Anti-Programmed Death 1 Monoclonal Antibody Dostarlimab for Patients With Recurrent or Advanced Mismatch Repair-Deficient Endometrial Cancer: A Nonrandomized Phase 1 Clinical Trial. JAMA Oncol. 2020, 6, 1766–1772. [Google Scholar] [CrossRef]
  73. Tang, Y.H.; Lin, C.Y.; Lai, C.H. Development of New Cancer Treatment by Identifying and Focusing the Genetic Mutations or Altered Expression in Gynecologic Cancers. Genes 2021, 12, 1593. [Google Scholar] [CrossRef]
Figure 1. Immunologic and genomic hallmarks of DEC. (A) DEC is not amenable to hormone therapy owing to its loss of ER and PR [5,6]. (B) Mutations in genes encoding subunits of the SWI/SNF chromatin remodeling complex could be targeted by small-molecule allosteric inhibitors and proteolysis-targeting chimeras (PROTACs) [29,60]. (C,D) Defects in MMR and the increased expression of PDL1 could be a target for immune checkpoint inhibitors [8,29,69,72]. (E) Mutations in FGFR2 and the amplification of CCNE2 could be a target for lenvatinib and Palbociclib [55]. (F) Mutation in TP53 could be a target for APE-246 and Wee1 kinase inhibitor [73]. For each inhibitor against the gene, the rationale of cancer treatment is described (signified by blue words).
Figure 1. Immunologic and genomic hallmarks of DEC. (A) DEC is not amenable to hormone therapy owing to its loss of ER and PR [5,6]. (B) Mutations in genes encoding subunits of the SWI/SNF chromatin remodeling complex could be targeted by small-molecule allosteric inhibitors and proteolysis-targeting chimeras (PROTACs) [29,60]. (C,D) Defects in MMR and the increased expression of PDL1 could be a target for immune checkpoint inhibitors [8,29,69,72]. (E) Mutations in FGFR2 and the amplification of CCNE2 could be a target for lenvatinib and Palbociclib [55]. (F) Mutation in TP53 could be a target for APE-246 and Wee1 kinase inhibitor [73]. For each inhibitor against the gene, the rationale of cancer treatment is described (signified by blue words).
Ijms 23 03794 g001
Table 1. Clinicopathologic features of UDEC.
Table 1. Clinicopathologic features of UDEC.
AuthorCase Number/UEC or DECSWI/SNF Deficiency (%)ARID1A/1B Co-DeficientMMR-d/-p (%)POLE Mutated/WTPDL1 +/− (%)p53 +/−Note
Hacking et al., 2019 [8]14/
4 UEC
10 DEC
NANA8/6 (57.1%)NA7/7 (50.0%)NAPDL1 not expressed in MMR-p tumors
Tessier-Cloutier et al., 2021 [11]82/
53 DEC
29 UEC
56 SWI/SNF deficiency (68.3%)25 (44.6%)38/18 (67.9%)2/54 (3.6%)NANA
26 SWI/SNF intact (31.7%)0 (0)12/14 (46.2%)4/22 (15.4%)NANA
Al-Hussaini et al., 2018 [18]17/
8 UEC- pure
4 UEC-mixed
5 DEC
BRG1/SMARCA4 loss: 3 a (20.0%)NA11/6 (64.7%)NA10/7 (66.7%)8/7 a
(53.3%)
a Two patients not applicable
Hamilton et al., 2022 [19]52/
17 UEC
35 DEC
34/15 b
SWI/SNF deficiency (69.4%)
22/27 b ARID1A/1B co-deficient (44.9%)30/13 c (69.8%)NANANAb 49 patients checked
c 43 patients checked
Hoang et al., 2016 [20]35 DEC20 Loss of BRG1 or INI1 (57.1%)NA13/7
(65.0%)
NANA1/19 (5.0%)
15 BRG1/INI1 intact (42.9%)NA11/4 (73.3%)NANA3/12 (20.0%)
Busca et al., 2020 [21]18 DEC/
11DEC-HG
7DEC-LG
11 DEC-HG3/6 (33.3%) (BRG1 loss/intact)NA6/5 (54.5%)NANA5/6 (45.4%)
7 DEC-LG1/4(20.0%)
(BRG1 loss/intact)
4/2 (66.6%) 0/5 (0)
Abbreviations: MMR-d, mismatch repair-deficient; MMR-p, mismatch repair-proficient; NA, not applicable. a although total patients is 17, the ratio was counted with 3/15 due to two patient’s data not applicable. b total patients is 52, the ratio was counted with 34/49 due to 49 patients’ data available. c total patients were 52, but there were 43 patients’ data available thus ratio counted with 30/43.
Table 2. Reported treatment in case series and clinical outcomes in UDEC.
Table 2. Reported treatment in case series and clinical outcomes in UDEC.
AuthorCase NumberDiagnosis Age (Mean, y-o)Stage (Cases)Treatment (Cases)DFS
(Months)
OS
(Months)
Outcome
Tessier-Cloutier et al., 2021 [11]8261SWI/SNF-deficient
I (22)
II (3)
III (15)
IV (16)
NANA2-year DSS for stage I–II: 65%
Stage III–IV: 3%
DOD (38)
NED (18)
64SWI/SNF-intact
I (13)
II (2)
III (7)
IV (4)
NANA2-year DSS for stage I–II: 100%
Stage III–IV: 61%
DOD (10)
AWD (1)
DOOC (2)
NED (13)
Busca et al., 2020 [21].1868DEC-HG
I–II (4)
III–IV (7)
Surgery (11)
CT (7)
RT (9)
1.7
2.1
10.7
NA
NA
NA
37.7
21.7
REC (2)
DOD (2)
NED (7)
72DEC-LG
I–II (6)
III–IV (1)
Surgery (7)
CT (3)
RT (7)
6.3 (recurrent patient)17+ (recurrent patient)REC (1)
NED (6)
Goh et al., 2020 [45]756II (1)
III (5)
IV (1)
Surgery + TP × 6+RT
Surgery + TP or TC × 5 – 6
NACT
15
58, 9, 5, 1, 2
0
15+
58+, 25, 21+, 6, NA
9
DOD (3)
AWD (1)
NED (2)
NA (1) *
Pfaendler et al., 2019 [49]256I (1)
III (1)
Surgery + TP × 6
Surgery + TP × 3
6
4
7
NA
DOD (1)
NA (1) *
Han et al., 2017 [50]461IA (1)
II (2)
IIIB (1)
None (1)
Pt refuse (1), RT (1)
CT + RT (1)
NA
1, NA
1
19+
7 weeks, 39+
10
DOD (2)
NED (2)
Silva et al., 2006 [51]2551 (median)I (14)
II (1)
III (6)
IV (4)
Surgery (24)
CT (18)
RT (4)
NA7
6–8+
DOD (15)
AWD (6)
NA (3)
Abbreviations: TP, paclitaxel + carboplatin; TC, paclitaxel + cisplatin; DFS, disease-free survival; OS, overall survival; RT, radiotherapy; CT, chemotherapy; NA, not applicable; REC, recurrence; DOD, died of disease; NED, no evidence of disease; AWD, alive with disease; DOOC, died of other cause; DSS, disease-specific survival; y-o, year-old; * move to other country/loss to follow up.
Table 3. Potential targets and existing inhibitors or activators in UDEC.
Table 3. Potential targets and existing inhibitors or activators in UDEC.
MechanismSubunit/Genetic TargetMedicationReference
Mismatch repair deficiency
PDL1/PD1 pathway
Anti-PD1 antibodyPembrolizumab[69]
Anti-PD1 antibodyDostarlimab[72]
Anti-FGFR2 BGJ398, infigratinib.[54]
ARID1AAurora A Alisertib[33]
SWI/SNF-polycomb antagonism PCR2, EZH2Tazemetostat[33]
SWI/SNF deficiencySynthetic lethal interactionInhibitors[33]
SMARCA4CDK4/6Palbociclib, abemaciclib, ribociclib
Aurora ATozasertib, alisertib
ARID1APARPTalazoparib, olaparib, rucaparib, veliparib
Abl, Src, c-KITDasatinib
SMARCB1HDACPanobinostat
UBE2CIxazomib, bortezomib
TP53 mutation
Mutant p53 cysteine residueAPR246[73]
Wee1Adavosertib[73]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Tung, H.-J.; Wu, R.-C.; Lin, C.-Y.; Lai, C.-H. Rare Subtype of Endometrial Cancer: Undifferentiated/Dedifferentiated Endometrial Carcinoma, from Genetic Aspects to Clinical Practice. Int. J. Mol. Sci. 2022, 23, 3794. https://doi.org/10.3390/ijms23073794

AMA Style

Tung H-J, Wu R-C, Lin C-Y, Lai C-H. Rare Subtype of Endometrial Cancer: Undifferentiated/Dedifferentiated Endometrial Carcinoma, from Genetic Aspects to Clinical Practice. International Journal of Molecular Sciences. 2022; 23(7):3794. https://doi.org/10.3390/ijms23073794

Chicago/Turabian Style

Tung, Hsiu-Jung, Ren-Chin Wu, Chiao-Yun Lin, and Chyong-Huey Lai. 2022. "Rare Subtype of Endometrial Cancer: Undifferentiated/Dedifferentiated Endometrial Carcinoma, from Genetic Aspects to Clinical Practice" International Journal of Molecular Sciences 23, no. 7: 3794. https://doi.org/10.3390/ijms23073794

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop