Next Article in Journal
The Microbiome and Genitourinary Cancers: A New Frontier
Previous Article in Journal
RBM39 Contributes to MGMT Maintenance in Response to Temozolomide-Induced DNA Damage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Association Between MMR Status and Prognostic Pathological Factors in Endometrioid Endometrial Cancer—A Single-Center Retrospective Study

by
Cezary Miedziarek
1,2,*,
Hubert Bochyński
1,
Katarzyna Bociańska
1,
Michał Potograbski
1,
Piotr Tyburski
1,
Mikołaj Piotr Zaborowski
1,3 and
Ewa Nowak-Markwitz
1
1
Division of Gynaecological Oncology, Department of Gynaecology, Poznan University of Medical Sciences, 61-701 Poznań, Poland
2
Doctoral School, Poznan University of Medical Sciences, 61-701 Poznań, Poland
3
Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-772 Poznań, Poland
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(22), 3605; https://doi.org/10.3390/cancers17223605 (registering DOI)
Submission received: 10 October 2025 / Revised: 27 October 2025 / Accepted: 31 October 2025 / Published: 8 November 2025
(This article belongs to the Section Cancer Pathophysiology)

Simple Summary

Clinical and pathological heterogeneity of endometrial carcinoma is not fully investigated, particularly in relation to mismatch repair (MMR) status. In practice, tumor profiles combining dMMR with adverse features may support therapy escalation, whereas favorable features support de-escalation. Identifying dMMR tumors has therapeutic relevance, guiding the consideration of immune checkpoint inhibitors. Previous studies have reported various findings regarding the impact of MMR deficiency, especially with respect to clinical progression of the disease, tumor grade, and the presence of adverse histopathological features such as lymphovascular space invasion (LVSI). By investigating these associations in our retrospective patient cohort, we aim to clarify the impact of MMR status on disease aggressiveness and to contribute evidence that may guide clinical decision-making. This study is centered on histopathological risk factors of endometrial cancer.

Abstract

Background/Objectives: Prognostic assessment in endometrial cancer (EC) is based on clinical and pathological features such as histological type, FIGO stage, tumor grade, LVSI, P53 status, and hormone receptor expression. Recent molecular research has distinguished four EC subtypes, with MMR status (pMMR vs. dMMR) providing clinically relevant stratification due to its predictive value for immunotherapy. The present study aims to compare dMMR and pMMR tumors in terms of the prevalence of adverse histopathological prognostic factors. Methods: This retrospective study included 179 patients with endometrioid endometrial carcinoma (EEC) treated at the authors’ institution (1 January 2023–31 August 2025). Patients were classified by MMR status (pMMR vs. dMMR) based on immunohistochemistry, and clinicopathological variables, including FIGO stage, myometrial invasion depth, tumor grade, LVSI, ER/PR expression, and P53 status, were analyzed. Normality was assessed using the Shapiro–Wilk test. Categorical variables were tested with chi-square or Fisher’s exact tests, reporting odds ratios with 95% CI, while continuous variables were compared using the Mann–Whitney test and presented as median (IQR) with the Hodges–Lehmann difference and 95% CI. Multivariable logistic regression with Wald tests was performed. Results: dMMR tumors accounted for 29.05% of all cases. Patients in the dMMR group were significantly more likely to present with FIGO stage III/IV disease (p = 0.036) and to exhibit LVSI (p = 0.008). No differences were observed between the groups with respect to tumor grade, estrogen receptor positivity, progesterone receptor positivity, or the prevalence of deep myometrial invasion. The most frequent pattern of protein loss in the dMMR population was concurrent loss of MLH1 and PMS2. Conclusions: In the studied population, dMMR tumors more frequently exhibited adverse prognostic features of EC, such as advanced stage of disease and lymphovascular space invasion. This suggests the potential for effective immunotherapy in this patient group.

1. Introduction

Endometrial cancer (EC) is a malignancy with a favorable prognosis. Considering all patients, the 5-year survival rate for this cancer is approximately 81% [1]. Prognostic assessment in EC depends on multiple clinical and pathological factors. The most important prognostic factors include histological tumor type, clinical stage of the disease, tumor grade, presence of lymphovascular space invasion (LVSI), and the presence of P53 gene mutations. Estrogen receptors (ER) and progesterone receptors (PR) positivity generally tracks with favorable biology and disease course, whereas receptor loss often coexists with adverse features [2,3].
The outcome of recent research on EC has been the identification of molecular subtypes of the disease, with practical implications. Based on The Cancer Genome Atlas (TCGA) analysis, EC has been molecularly classified into four subtypes: POLE ultramutated, microsatellite instability-high (MSI-H), copy-number low (CN-L), and copy-number high (CN-H) [4]. Accordingly, major guidelines incorporate molecular subtype into postoperative risk stratification and adjuvant therapy decisions (e.g., potential de-escalation for POLE ultramutated and escalation for TP53-mutated) [5]. Microsatellites are short repeats composed of a few nucleotides that are distributed throughout the genome. During DNA replication, errors can occur within these repeats. In normal cells, such errors are corrected by the DNA mismatch repair (MMR) system. When MMR is intact (pMMR), microsatellite length is preserved. When MMR is deficient (dMMR), errors accumulate, leading to microsatellite instability (MSI). In EC, tumors are therefore commonly classified as pMMR or dMMR, a distinction with important clinical implications, as dMMR/MSI-high tumors are particularly responsive to immune checkpoint inhibition [6,7,8]. Current guidelines endorse universal tumor testing for MMR deficiency in endometrial cancer, typically by MMR immunohistochemistry (IHC) with MSI testing (PCR or NGS) as an alternative or confirmatory approach [9]. On IHC, loss of MSH2, MSH6, or isolated PMS2 strongly suggests Lynch syndrome (LS) and directs germline testing toward the corresponding MMR gene, whereas MLH1 may also suggest sporadic MLH promoter hypermethylation (MLH1-PHM) with transcriptional silencing. Rare LS-like cases due to constitutional MLH1 methylation have been reported, but with limited need for genetic counseling, because they are not associated with heritable (germline) alterations [10]. Evidence linking MMR status to histopathologic aggressiveness in EC, including FIGO stage, LVSI, grade, and depth of invasion, remains inconsistent across studies. This knowledge gap motivates the comparison of pMMR and dMMR tumors using routinely reported pathologic features.
MMR status is a recognized predictor of response to immunotherapy in EC [6]. Consequently, scientific studies increasingly consider dMMR and pMMR populations as two distinct entities, requiring separate investigations due to their differences in treatment response. We hypothesized that dMMR tumors exhibit more aggressive pathological features, as they entail high mutational burden and a dysregulated immune microenvironment, alongside genomic instability [1,2,5]. The aim of the present study is to compare dMMR and pMMR EC patients with respect to the prevalence of pathological prognostic factors.

2. Materials and Methods

This retrospective study included patients treated for EC over a two-year period (1 January 2023–31 August 2025) at the authors’ affiliated institution. We enrolled patients consecutively, applying prespecified exclusions (non-endometrioid histology, missing core variables). The study was conducted at the Department of Gynecologic Oncology, Poznan University of Medical Sciences. The Bioethics Committee of Poznan University of Medical Sciences confirmed that, due to the retrospective nature of the study, no additional approval was required (Confirmation KB-655/25). The study was conducted in accordance with the principles outlined in the Declaration of Helsinki. Details of patient selection are presented in the flow chart (Figure 1).
The study included patients who were diagnosed with EC. Among these, only those with an endometrioid histopathological subtype (EEC) were analyzed. Histopathological diagnoses based on biopsy and postoperative specimens, surgical descriptions, and imaging studies were reviewed. Patients were divided into two main groups according to their MMR status, determined by immunohistochemical testing for the loss of expression of the MLH1, PMS2, MSH2, and MSH6 proteins: pMMR (proficient MMR) and dMMR (deficient MMR). Focal MMR loss was considered noninterpretable. We relied on unequivocally stained regions and repeated IHC as needed. Reproducible focal loss suggestive of clonal deficiency was flagged as suspicious for dMMR, prompted genetic testing [11], and revealed MSI-high in all such cases (n = 8). Patient data regarding tumor stage according to FIGO were collected, and for analysis purposes, patients were categorized into two groups: those with stage I and II disease and those with stage III and IV disease. Independently, patients with deep (≥50%) and superficial (<50%) myometrial invasion were identified. Tumors were also classified by grade into low-grade (G1 and G2) and high-grade (G3). LVSI status was considered positive if it was substantial or focal, and negative if it was absent. ER and PR status were defined according to the Allred score, where scores 0–2 were considered negative and scores 3–8 were considered positive [12]. Wild-type P53 (P53-wt) was defined as heterogeneous or variable nuclear staining with an internal control present. Aberrant (P53-abn) was defined by diffuse strong nuclear overexpression, complete absence of nuclear staining (null) with intact internal controls, or cytoplasmic staining with absent nuclear staining [13,14]. Mutation analysis of the POLE and TP53 genes was not performed using molecular methods, as these tests were not yet implemented in routine practice at the whole time of data collection. Additionally, in our center, immunohistochemical results for P53 generally correlate well with genetic mutation analysis. Antibodies used for ICH are presented in Table 1.
MSI assays were not available for all dMMR cases, particularly those from the early data collection period. However, in the tested cases (n = 34), all tumors were found to be MSI-high. Non-standard immunohistochemical patterns (e.g., isolated loss of a single MMR protein) prompted expedited referral for genetic counseling and evaluation for Lynch syndrome, as per institutional practice. These data are not included in the present study, which focuses on histopathologic determinants. It has to be noted that, as MSI assays were not available for all tumors and MMR status was determined by IHC, this approach can misclassify a minority of cases, e.g., those with non-standard immunohistochemical patterns.
Statistical analysis, including both descriptive and comparative methods, was conducted using Microsoft Excel and RStudio 2024.12.1. Group comparisons for categorical variables used chi-square or Fisher’s exact tests, as appropriate. For 2 × 2 tables, we additionally reported odds ratios (OR) with 95% confidence intervals calculated by Fisher’s exact method. Continuous variables were compared using the Mann–Whitney test and are summarized as median (IQR) with Hodges–Lehmann differences and 95% CI. Observations with missing data were excluded from bivariate analyses. We performed multivariable analyses for FIGO and LVSI (multivariable logistic regression with Wald tests). For the FIGO analysis, we excluded depth of myometrial invasion and LVSI, as these are elements of the staging definition. ER and PR status were also excluded due to substantial missing data and a significantly different proportion of missing values between the dMMR and pMMR groups (p = 0.0287, chi-square). For the LVSI analysis, we excluded FIGO (the FIGO classification incorporates LVSI) and ER and PR status due to the high proportion of missing data. We prespecified covariates and did not use stepwise selection. Complete case analysis, ORs with 95% CIs, and AUC are reported.
Missing data were handled by complete-case analysis for all inferential tests and models. ‘Unknown’ values were used only for descriptive summaries. We prespecified this approach to avoid ad hoc imputation and because of high rate of missingness for ER/PR data.

3. Results

The study group consists of 179 female patients (mean age 63.76 ± 10.43) with EC of the endometrioid histological subtype treated at our clinic. Selected epidemiologic factors—age, body weight and BMI—are summarized in Table 2. A total of 127 (70.95%) were classified as pMMR and 52 (29.05%) as dMMR. A detailed comparison of the two groups with respect to the analyzed parameters is presented in Table 3. The category “Unknown” is included for descriptive purposes only. For the analysis, we employed complete-case analysis, excluding observations with missing values in any variable included in a given model.
In bivariate tests, patients in the dMMR group were significantly more likely to be LVSI-positive compared with the pMMR group. Patients with dMMR tumors more frequently presented with FIGO stage III–IV disease. The groups did not differ significantly in histological grade, P53 status, estrogen receptor positivity, progesterone receptor positivity, and the prevalence of deep myometrial invasion. The groups were also comparable in terms of age, body weight, and BMI.
In a multivariable model (outcome: dMMR vs. pMMR) including FIGO, LVSI, grade, P53, and myoinvasion, FIGO III/IV was not significant: aOR = [1.95] (95% CI [0.72–5.25], p = 0.187). LVSI remained significant: aOR = [2.96] (95% CI [1.25–7.04], p = 0.014). It has to be noted, that the model showed modest discrimination (AUC = [0.646], 95% CI [0.546–0.735]) (Table 4).
In multivariate analysis, the presence of LVSI was independently associated with dMMR status (aOR [3.38], 95% CI [1.46–7.81]; p = 0.004). Additionally, it was associated with deep myometrial invasion (aOR [12.45], 95% CI [5.15–30.11]; p = <0.001, model discrimination: AUC = [0.807] (95% CI [0.743–0.867]). Advanced stage (FIGO III/IV) was independently associated with dMMR status (aOR [2.68], 95% CI [1.11–6.47]; p = 0.028). Additionally, it was associated with high histological grade (aOR [3.85], 95% CI [1.45–10.19]; p = [0.007]). Model discrimination: AUC = [0.709] (95% CI [0.604–0.811]) (Table 5 and Table 6).
An additional analysis was performed to evaluate the combinations of mismatch repair protein loss associated with dMMR. The most common pattern of absent expression on immunohistochemistry was combined loss of MLH1 and PMS2, which occurred in 78.8% of cases (Table 7).

4. Discussion

The dMMR population accounted for 29.05% of patients, which is consistent with previous reports indicating that this alteration occurs in approximately 25–30% of ECs [15,16]. It is worth noting that these analyses encompass all histopathological subtypes of EC. Nevertheless, the endometrioid subtype remains the most prevalent.
Data regarding the impact of MMR status on the clinical and pathological characteristics of EC remain inconclusive [17]. In our studied population, patients with EC and dMMR status were significantly more likely to present with a higher FIGO clinical stage and exhibited LVSI more frequently. It has to be noted that our results are associations and do not imply causation. Several studies have demonstrated an association between MMR status and the clinical stage of EC at diagnosis, where dMMR endometrial carcinomas are considered to exhibit a more aggressive clinical course [18]. In these studies, patients with dMMR EC are considered more likely to present with FIGO stage III or IV disease [19,20,21]. Moreover, they tend to have larger tumor size and a higher incidence of lymph node involvement, both of which are associated with advanced FIGO classification [19]. Reports also indicate a significantly lower prevalence of FIGO stage I disease among dMMR EC compared with pMMR tumors, with a relative shift toward stages II–IV [22]. On the other hand, several large studies have suggested that dMMR EC is more often present at lower tumor stages compared with MMR-proficient cases. Nonetheless, these reports consistently highlight that the dMMR subgroup more frequently exhibits metastatic parameters such as LVSI [17,23,24]. Given that dMMR cancers are more often associated with adverse clinicopathologic features, the implementation of immunotherapy is particularly valuable, especially in patients with advanced disease, as this subgroup derives the most significant benefit from immune checkpoint inhibition [18,25,26]. It is worth noting that differences in FIGO stage across studies may reflect heterogeneity in patient selection and IHC protocols, as well as variations in reporting schemes (e.g., stage I reported separately vs. pooling I + II as ‘early’ and III + IV as ‘advanced’). Differences in ethnic composition across referenced studies and the monoethnic nature of our cohort are also relevant to between-population comparisons.
LVSI is defined as the presence of malignant cells within lymphatic and/or vascular spaces. It represents an adverse prognostic factor in EC, being associated with lymph node metastasis, an increased risk of pelvic recurrence, and reduced overall survival. The impact is significant for both 3-year and 5-year survival rates, with the latter decreasing by 13–25% in LVSI-positive disease [27]. Women with dMMR tumors were more likely to have higher-grade cancers and more frequent LVSI [28], the latter consistent with our results.
Available analyses provide conflicting evidence regarding the association between tumor grade and MMR status. Some studies have demonstrated that high-grade tumors occur more frequently in the dMMR population, whereas others report them to be less common or similarly distributed, as observed in our cohort [28,29,30]. This inconsistency highlights the need for additional population-based studies in this area, particularly since tumor grade continues to be a key factor influencing therapeutic decision-making in early-stage disease [31].
Studies emphasize that in the majority of EEC, including dMMR tumors, estrogen and progesterone receptors are expressed, and P53 displays a wild-type pattern [32,33]. This is consistent with the findings of our analysis. Positive expression of estrogen and progesterone receptors, as well as the absence of TP53 mutations, represent favorable prognostic factors [34,35]. High-grade tumors constitute approximately 15–20% of endometrial cancers when a binary grading scheme is used. They are associated with worse clinical outcomes and more frequent adverse molecular alterations, including TP53 mutations [2]. The relationship between grade and the prevalence of ER/PR positivity remains controversial and varies across studies [34].
Additional analysis of immunohistochemistry patterns in dMMR EC cells reveals that the most prevalent phenotype is the loss of expression of both MLH1 and PMS2 proteins, accounting for approximately 70–80% of cases, which is in agreement with the outcomes from our own cohort. This reflects the instability of the PMS2 protein in the absence of its MLH1 partner [36,37]. The MSH2 and MSH6 co-loss is the second most common combination (15–20%), typically indicative of MSH2 mutations [26]. Less frequently, isolated MSH6 or PMS2 losses occur (5–10%), corresponding to mutations in the respective genes [38,39]. Interestingly, in our patient population, the second most common finding was isolated loss of single proteins, namely MSH6, MLH1, or MSH2, not MSH2 and MSH6 co-loss, which is considered a less common situation [27].

5. Limitations

A limitation of our study is the lack of genetic testing for POLE and TP53 mutations. The presence of POLE mutation represents an independent favorable prognostic factor in patients with endometrial carcinoma [40]. Genetic testing for TP53 mutations was also not included, as genetic assays were introduced into clinical practice after the initiation of data collection for this study. Furthermore, immunohistochemistry has been shown to correlate well with genetic testing for TP53 [41,42,43]. Therefore, it is unlikely that the omission of these analyses would have significantly affected our results.
However, in clinical practice, without POLE and TP53 sequencing, residual TCGA misclassification is possible. Favorable POLE-mutant and poor-prognosis TP53-aberrant tumors may be grouped solely based on clinical and pathological factors. While this analysis relied on IHC surrogates to show histologic features, full molecular profiling, including POLE and TP53 genetic testing, will be undertaken for any patient for whom it is expected to change clinical decision-making. This is important because POLE-ultramutated tumors (even with adverse histology such as LVSI or deep invasion) generally have an excellent prognosis and may warrant de-escalation of adjuvant therapy, whereas TP53-mutated tumors carry an unfavorable prognosis and often justify treatment escalation.
Although MMR IHC generally aligns well with MSI-high status in endometrial cancer, numerous studies have documented discordant cases. Therefore, the absence of MSI testing in every case should be regarded as a limitation.
As a single-center study, our cohort may reflect referral bias due to the case mix specific to our institution. It is limited by sample size, which may reduce precision and the generalizability of estimates. The benefit of a single-center design is histopathological consistency and uniform IHC.

6. Conclusions

EECs differ in their clinical and histopathological phenotypes depending on MMR status. Our findings are in line with reports indicating that dMMR tumors are characterized by a more aggressive clinical course and a higher prevalence of adverse histopathological features. In our cohort, this was reflected by a greater frequency of advanced FIGO stage and more common occurrence of LVSI in the dMMR group. These observations underscore the importance of adjuvant treatment, particularly immunotherapy, which has proven to be especially effective in this subset of patients. Future multicenter studies should integrate MMR status with comprehensive molecular profiling (POLE and TP53) to refine risk stratification and guide adjuvant therapy. Clinically, dMMR is a validated predictive biomarker for response to immune checkpoint inhibition, while LVSI is an established adverse prognostic feature. Together, these variables could support risk-adapted enrichment in immunotherapy trials, e.g., prioritizing dMMR tumors and considering dMMR with LVSI as a high-risk disease. Such strategies may increase event rates and therapeutic signal detection while aligning with routine pathology workflows. Prospective, biomarker-driven trials with standardized staging and pathology review are needed to confirm their benefits.

Author Contributions

Conceptualization, C.M., H.B., E.N.-M., M.P.Z. and K.B.; methodology, C.M., M.P.Z. and E.N.-M.; software, C.M.; formal analysis, E.N.-M.; investigation, C.M., H.B., K.B., P.T., M.P. and M.P.Z.; resources, C.M., H.B., P.T., M.P., M.P.Z. and E.N.-M.; writing—original draft preparation, C.M., H.B. and K.B.; writing—review and editing, C.M., M.P.Z. and E.N.-M.; visualization, C.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was co-funded by: the Polish Minister of Science and Higher Education within the program ‘Studenckie Koła Naukowe Tworzą Innowacje’ (Student Scientific Associations Create Innovations) grant number SKN/SP/602358/2024; co-funded from the large research grant from statutory funding for young researchers—PUMS doctoral students for 2025 NMN0000119. The APC was funded by: The Vice-Rector for Research’s (Prorektor ds. Nauki) budget of the Poznan University of Medical Sciences. The budget of the Council of Scientific Colleges (Rada Kolegiów Nauk) of the Poznan University of Medical Sciences in Poznań. Development Foundation (Fundacja Rozwoju) of the Poznan University of Medical Sciences. Statutory funds of the author’s affiliation centers.

Institutional Review Board Statement

Ethical review and approval were waived for this study due to the decision of Poznan University of Medical Sciences Ethics Committee No KB-655/25.

Informed Consent Statement

Written informed consent was waived for this study due to the decision of Poznan University of Medical Sciences Ethics Committee No KB-655/25.

Data Availability Statement

Data are contained within the article.

Acknowledgments

We would like to thank the students of the Student Research Group at our Clinic for their assistance with data collection: Magdalena Nadolna, Jędrzej Sikora, Marta Szczygieł, Mateusz Szot, Stefan Malewski.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tejerizo-García, Á.; Jiménez-López, J.S.; Muñoz-González, J.L.; Bartolomé-Sotillos, S.; Marqueta-Marqués, L.; López-González, G.; Gómez, J.F.P.-R. Overall Survival and Disease-Free Survival in Endometrial Cancer: Prognostic Factors in 276 Patients. OncoTargets Ther. 2013, 6, 1305–1313. [Google Scholar] [CrossRef] [PubMed]
  2. Coll-de la Rubia, E.; Martinez-Garcia, E.; Dittmar, G.; Gil-Moreno, A.; Cabrera, S.; Colas, E. Prognostic Biomarkers in Endometrial Cancer: A Systematic Review and Meta-Analysis. J. Clin. Med. 2020, 9, 1900. [Google Scholar] [CrossRef]
  3. Wang, X.; Aziz, A.u.R.; Wang, D.; Wang, Y.; Liu, M.; Yu, X.; Wang, D. Prognostic Factors and Survival Outcomes of Immunohistochemically Detection Based-Molecular Subtypes of Endometrial Cancer–Analysis of 576 Clinical Cases. Diagn. Pathol. 2024, 19, 162. [Google Scholar] [CrossRef]
  4. Cancer Genome Atlas Research Network; Kandoth, C.; Schultz, N.; Cherniack, A.D.; Akbani, R.; Liu, Y.; Shen, H.; Robertson, A.G.; Pashtan, I.; Shen, R.; et al. Integrated Genomic Characterization of Endometrial Carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef]
  5. Galant, N.; Krawczyk, P.; Monist, M.; Obara, A.; Gajek, Ł.; Grenda, A.; Nicoś, M.; Kalinka, E.; Milanowski, J. Molecular Classification of Endometrial Cancer and Its Impact on Therapy Selection. Int. J. Mol. Sci. 2024, 25, 5893. [Google Scholar] [CrossRef]
  6. Corr, B.; Cosgrove, C.; Spinosa, D.; Guntupalli, S. Endometrial Cancer: Molecular Classification and Future Treatments. BMJ Med. 2022, 1, e000152. [Google Scholar] [CrossRef]
  7. Adorisio, R.; Troncone, G.; Barberis, M.; Pepe, F. Molecular Profiling of H-MSI/dMMR/for Endometrial Cancer Patients: “New Challenges in Diagnostic Routine Practice”. J. Mol. Pathol. 2024, 5, 187–198. [Google Scholar] [CrossRef]
  8. Paranjpe, R.; Chen, C.; Sun, Y.; Hong, J.; Prabhu, V.S.; Ojha, J.; Meng, R.; Duska, L.R. Molecular Heterogeneity in dMMR/MSI-H Endometrial Cancer in the US. J. Clin. Oncol. 2025, 43, e17643. [Google Scholar] [CrossRef]
  9. Concin, N.; Matias-Guiu, X.; Cibula, D.; Colombo, N.; Creutzberg, C.L.; Ledermann, J.; Mirza, M.R.; Vergote, I.; Abu-Rustum, N.R.; Bosse, T.; et al. ESGO-ESTRO-ESP Guidelines for the Management of Patients with Endometrial Carcinoma: Update 2025. Lancet Oncol. 2025, 26, e423–e435. [Google Scholar] [CrossRef]
  10. Plotkin, A.; Olkhov-Mitsel, E.; Nofech-Mozes, S. MLH1 Methylation Testing as an Integral Component of Universal Endometrial Cancer Screening—A Critical Appraisal. Cancers 2023, 15, 5188. [Google Scholar] [CrossRef] [PubMed]
  11. Wang, C.; Zhang, L.; Vakiani, E.; Shia, J. Detecting Mismatch Repair Deficiency in Solid Neoplasms: Immunohistochemistry, Microsatellite Instability, or Both? Mod. Pathol. 2022, 35, 1515–1528. [Google Scholar] [CrossRef]
  12. Ahmad Fauzi, M.F.; Wan Ahmad, W.S.H.M.; Jamaluddin, M.F.; Lee, J.T.H.; Khor, S.Y.; Looi, L.M.; Abas, F.S.; Aldahoul, N. Allred Scoring of ER-IHC Stained Whole-Slide Images for Hormone Receptor Status in Breast Carcinoma. Diagnostics 2022, 12, 3093. [Google Scholar] [CrossRef]
  13. Armbruster, H.; Schotte, T.; Götting, I.; Overkamp, M.; Granai, M.; Volmer, L.L.; Bahlinger, V.; Matovina, S.; Koch, A.; Dannehl, D.; et al. Aberrant P53 Immunostaining Patterns in Breast Carcinoma of No Special Type Strongly Correlate with Presence and Type of TP53 Mutations. Virchows Arch. 2024, 485, 631–642. [Google Scholar] [CrossRef]
  14. Mcgregor, M.J.; Fadhil, W.; Wharton, R.; Yanagisawa, Y.; Presz, M.; Pritchard, A.; Womack, C.; Dutton, S.; Kerr, R.S.; Kerr, D.J.; et al. Aberrant P53 Expression Lacks Prognostic or Predictive Significance in Colorectal Cancer: Results from the VICTOR Trial. Anticancer Res. 2015, 35, 1641–1645. [Google Scholar]
  15. Lorenzi, M.; Amonkar, M.; Zhang, J.; Mehta, S.; Liaw, K.-L. Epidemiology of Microsatellite Instability High (MSI-H) and Deficient Mismatch Repair (dMMR) in Solid Tumors: A Structured Literature Review. J. Oncol. 2020, 2020, 1807929. [Google Scholar] [CrossRef]
  16. Slomovitz, B.M.; Cibula, D.; Lv, W.; Ortaç, F.; Hietanen, S.; Backes, F.; Kikuchi, A.; Lorusso, D.; Dańska-Bidzińska, A.; Samouëlian, V.; et al. Pembrolizumab or Placebo Plus Adjuvant Chemotherapy With or Without Radiotherapy for Newly Diagnosed, High-Risk Endometrial Cancer: Results in Mismatch Repair-Deficient Tumors. J. Clin. Oncol. 2025, 43, 251–259. [Google Scholar] [CrossRef]
  17. Jumaah, A.S.; Al-Haddad, H.S.; Salem, M.M.; McAllister, K.A.; Yasseen, A.A. Mismatch Repair Deficiency and Clinicopathological Characteristics in Endometrial Carcinoma: A Systematic Review and Meta-Analysis. J. Pathol. Transl. Med. 2021, 55, 202–211. [Google Scholar] [CrossRef] [PubMed]
  18. Marchetti, M.; Ferrari, J.; Vezzaro, T.; Masatti, L.; Tasca, G.; Maggino, T.; Tozzi, R.; Saccardi, C.; Noventa, M.; Spagnol, G. The Role of Immunotherapy in MMR-Deficient Endometrial Carcinoma: State of the Art and Future Perspectives. J. Clin. Med. 2024, 13, 7041. [Google Scholar] [CrossRef] [PubMed]
  19. Ring, K.L.; Connor, E.V.; Atkins, K.A.; Ricketts, W.; Kashlan, B.; Modesitt, S.C. Women 50 Years or Younger with Endometrial Cancer: The Argument for Universal Mismatch Repair Screening and Potential for Targeted Therapeutics. Int. J. Gynecol. Cancer 2013, 23, 853–860. [Google Scholar] [CrossRef]
  20. Gordhandas, S.; Kahn, R.M.; Gamble, C.; Talukdar, N.; Maddy, B.; Nelson, B.B.; Askin, G.; Christos, P.J.; Holcomb, K.; Caputo, T.A.; et al. Clinicopathologic Features of Endometrial Cancer with Mismatch Repair Deficiency. Ecancermedicalscience 2020, 14, 1061. [Google Scholar] [CrossRef] [PubMed]
  21. Kaneko, E.; Sato, N.; Sugawara, T.; Noto, A.; Takahashi, K.; Makino, K.; Terada, Y. MLH1 Promoter Hypermethylation Predicts Poorer Prognosis in Mismatch Repair Deficiency Endometrial Carcinomas. J. Gynecol. Oncol. 2021, 32, e79. [Google Scholar] [CrossRef] [PubMed]
  22. Pasanen, A.; Loukovaara, M.; Bützow, R. Clinicopathological Significance of Deficient DNA Mismatch Repair and MLH1 Promoter Methylation in Endometrioid Endometrial Carcinoma. Mod. Pathol. 2020, 33, 1443–1452. [Google Scholar] [CrossRef] [PubMed]
  23. Hathout, L.; Sherwani, Z.K.; Alegun, J.; Ohri, N.; Fields, E.C.; Shah, S.; Beriwal, S.; Horne, Z.D.; Kidd, E.A.; Leung, E.W.; et al. Prognostic Effect of Mismatch Repair Status in Early-Stage Endometrial Cancer Treated With Adjuvant Radiation: A Multi-Institutional Analysis. Int. J. Radiat. Oncol. Biol. Phys. 2024, 119, 1158–1165. [Google Scholar] [CrossRef] [PubMed]
  24. Keverline, K.J.; Hill, B.; Hom-Tedla, M.; Jacobs, M.; Eskander, R.N. Impact of Mismatch Repair (MMR) Status on Recurrence in High Intermediate Risk Endometrial Cancer. Gynecol. Oncol. 2025, 197, 116–120. [Google Scholar] [CrossRef]
  25. Mahdi, H.; Chelariu-Raicu, A.; Slomovitz, B.M. Immunotherapy in Endometrial Cancer. Int. J. Gynecol. Cancer 2023, 33, 351–357. [Google Scholar] [CrossRef]
  26. Carvalho, F.M.; Carvalho, J.P. Unraveling the Heterogeneity of Deficiency of Mismatch Repair Proteins in Endometrial Cancer: Predictive Biomarkers and Assessment Challenges. Cancers 2024, 16, 3452. [Google Scholar] [CrossRef]
  27. Yarandi, F.; Shirali, E.; Akhavan, S.; Nili, F.; Ramhormozian, S. The Impact of Lymphovascular Space Invasion on Survival in Early Stage Low-Grade Endometrioid Endometrial Cancer. Eur. J. Med. Res. 2023, 28, 118. [Google Scholar] [CrossRef]
  28. Backes, F.J.; Haag, J.; Cosgrove, C.M.; Suarez, A.; Cohn, D.E.; Goodfellow, P.J. Mismatch Repair Deficiency Identifies Patients with High-Intermediate–Risk (HIR) Endometrioid Endometrial Cancer at the Highest Risk of Recurrence: A Prognostic Biomarker. Cancer 2019, 125, 398–405. [Google Scholar] [CrossRef]
  29. 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]
  30. Fountzilas, E.; Kotoula, V.; Pentheroudakis, G.; Manousou, K.; Polychronidou, G.; Vrettou, E.; Poulios, C.; Papadopoulou, E.; Raptou, G.; Pectasides, E.; et al. Prognostic Implications of Mismatch Repair Deficiency in Patients with Nonmetastatic Colorectal and Endometrial Cancer. ESMO Open 2019, 4, e000474. [Google Scholar] [CrossRef]
  31. Boer, S.M.d.; Powell, M.E.; Mileshkin, L.; Katsaros, D.; Bessette, P.; Haie-Meder, C.; Ottevanger, P.B.; Ledermann, J.A.; Khaw, P.; Colombo, A.; et al. Adjuvant Chemoradiotherapy versus Radiotherapy Alone for Women with High-Risk Endometrial Cancer (PORTEC-3): Final Results of an International, Open-Label, Multicentre, Randomised, Phase 3 Trial. Lancet Oncol. 2018, 19, 295–309. [Google Scholar] [CrossRef]
  32. Saharti, S.; Altaf, F. Mismatch Repair Proteins Evaluation in Endometrial Carcinoma: Clinicopathological Features and Prognostic Implications. Available online: https://www.ejgo.net/articles/10.22514/ejgo.2024.076?utm_source=chatgpt.com (accessed on 8 September 2025).
  33. Vrede, S.W.; Weelden, W.J.V.; Bulten, J.; Gilks, C.B.; Teerenstra, S.; Huvila, J.; Matias-Guiu, X.; Gil-Moreno, A.; Asberger, J.; Sweegers, S.; et al. Hormonal Biomarkers Remain Prognostically Relevant within the Molecular Subgroups in Endometrial Cancer. Gynecol. Oncol. 2025, 192, 15–23. [Google Scholar] [CrossRef]
  34. Yadav, A.; Sistla, A.; Swain, M.; Gowrishankar, S.; Padua, M.d.; Modi, T.; Himabindu, R.; Agarwal, N.; Kulkarni, A.; Bhandari, T.; et al. To Study the Expression of Estrogen, Progesterone Receptor and P53 Immunohistochemistry Markers in Subtyping Endometrial Carcinoma. Indian J. Pathol. Microbiol. 2024, 67, 62–67. [Google Scholar] [CrossRef] [PubMed]
  35. Wood, R. Optimizing Immunohistochemistry Reporting in Endometrial Cancer. Cureus 2024, 16, e65810. [Google Scholar] [CrossRef]
  36. Nádorvári, M.L.; Kenessey, I.; Kiss, A.; Barbai, T.; Kulka, J.; Rásó, E.; Tímár, J. Comparison of Standard Mismatch Repair Deficiency and Microsatellite Instability Tests in a Large Cancer Series. J. Transl. Med. 2024, 22, 150. [Google Scholar] [CrossRef]
  37. Aytekin, O.; Cesur, N.; Gozel, S.; Karsli, S.E.; Tokalioglu, A.A.; Kilic, F.; Cuylan, Z.F.; Selcuk, I.; Comert, G.K.; Erdogan, F.; et al. Mismatch Repair Protein Deficiency and Its Relationship with Clinicopathological Factors in Endometrial Cancer: A Retrospective Study. J. Cancer 2025, 16, 2778–2786. [Google Scholar] [CrossRef]
  38. Mismatch Repair Deficiency as a Predictive and Prognostic Biomarker in Endometrial Cancer: A Review on Immunohistochemistry Staining Patterns and Clinical Implications. Available online: https://www.mdpi.com/1422-0067/25/2/1056?utm_source=chatgpt.com (accessed on 7 September 2025).
  39. Lachit, K.; Vinita, P. Study of Mismatch Repair Protein Expression by Using Immunohistochemistry in Various Carcinomas with Special Reference to Colorectal Adenocarcinomas—At a Tertiary Referral Laboratory in India. Asian Pac. J. Cancer Biol. 2022, 7, 341–347. [Google Scholar] [CrossRef]
  40. Wu, Q.; Zhang, N.; Xie, X. The Clinicopathological Characteristics of POLE-Mutated/Ultramutated Endometrial Carcinoma and Prognostic Value of POLE Status: A Meta-Analysis Based on 49 Articles Incorporating 12,120 Patients. BMC Cancer 2022, 22, 1157. [Google Scholar] [CrossRef]
  41. Eich, M.-L.; Siemanowsk-Hrach, J.; Drebber, U.; Friedrichs, N.; Mallmann, P.; Domröse, C.; Ratiu, D.; Merkelbach-Bruse, S.; Büttner, R.; Quaas, A.; et al. Assessment of P53 in Endometrial Carcinoma Biopsy and Corresponding Hysterectomy Cases in a Real-World Setting: Which Cases Need Molecular Work-Up? Cancers 2025, 17, 1506. [Google Scholar] [CrossRef] [PubMed]
  42. Thiel, K.W.; Devor, E.J.; Filiaci, V.L.; Mutch, D.; Moxley, K.; Alvarez Secord, A.; Tewari, K.S.; McDonald, M.E.; Mathews, C.; Cosgrove, C.; et al. TP53 Sequencing and P53 Immunohistochemistry Predict Outcomes When Bevacizumab Is Added to Frontline Chemotherapy in Endometrial Cancer: An NRG Oncology/Gynecologic Oncology Group Study. J. Clin. Oncol. 2022, 40, 3289–3300. [Google Scholar] [CrossRef]
  43. Vermij, L.; Léon-Castillo, A.; Singh, N.; Powell, M.E.; Edmondson, R.J.; Genestie, C.; Khaw, P.; Pyman, J.; McLachlin, C.M.; Ghatage, P.; et al. P53 Immunohistochemistry in Endometrial Cancer: Clinical and Molecular Correlates in the PORTEC-3 Trial. Mod. Pathol. 2022, 35, 1475–1483. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow-chart presenting the selection of final cohort for MMR comparison.
Figure 1. Flow-chart presenting the selection of final cohort for MMR comparison.
Cancers 17 03605 g001
Table 1. Antibodies used for ICH. All immunohistochemical stains were performed on the DAKO Autostainer Link 48 using the EnVision™ FLEX, High pH detection system; all antibodies were supplied by DAKO.
Table 1. Antibodies used for ICH. All immunohistochemical stains were performed on the DAKO Autostainer Link 48 using the EnVision™ FLEX, High pH detection system; all antibodies were supplied by DAKO.
TargetAntibodyClone
P53Flex RTU Monoclonal Mouse Anti-Human, P53 ProteinDO-7
ERFlex RTU Monoclonal Rabbit Anti-Human, Estrogen Receptor alfaEP1
PRFlex RTU Monoclonal Mouse Anti-Human, Progesterone ReceptorPgR 636
MLH1Flex RTU Monoclonal Mouse Anti-Human, MutL Protein Homolog 1, MLH1ES05
PMS2Flex RTU Monoclonal Rabbit Anti-Human, Postmeiotic Segregation Increased 2, PMS2EP51
MSH2Flex RTU Monoclonal Mouse Anti-Human, MutS Protein Homolog 2, MSH2FE11
MSH6Flex RTU Monoclonal Rabbit Anti-Human, MutS Protein Homolog 6, MSH6EP49
Table 2. Selected epidemiologic factors summary. HL—Hodges–Lehmann differences; CI—confidence interval.
Table 2. Selected epidemiologic factors summary. HL—Hodges–Lehmann differences; CI—confidence interval.
VariableTotal (n = 179)pMMR (n = 127)dMMR (n = 52)p-ValueHL95% CI
Age [years]
mean ± SD
63.76 ± 10.4363.31 ± 10.2164.85 ± 10.970.2552.00−1.00–5.00
Body weight [kg]
median, IQR
77 (67–93)77 (67–93)76.5 (68–92)0.7091.00−7.00–4.00
BMI [kg/m2]
median, IQR
29.67 (25.71–34.85)29.62 (25.86–34.29)30.18 (25.06–35.3)0.868−0.19−2.55–1.89
Table 3. Study group characteristics. The designation ‘unknown’ in the myometrial invasion section refers to cases where the available material was insufficient for assessment. P53 and hormone receptor status were not determined in specimens with only superficial myometrial infiltration and limited tissue. OR—odds ratio; CI—confidence interval.
Table 3. Study group characteristics. The designation ‘unknown’ in the myometrial invasion section refers to cases where the available material was insufficient for assessment. P53 and hormone receptor status were not determined in specimens with only superficial myometrial infiltration and limited tissue. OR—odds ratio; CI—confidence interval.
VariableTotal (n = 179)pMMR (n = 127)dMMR (n = 52)p-ValueOR95% CI
FIGO 0.036 *2.631.04–6.63
-
I/II
150 (83.80%)111 (87.40%)39 (75.00%)
-
III/IV
27 (15.08%)14 (11.02%)13 (25.00%)
-
Unknown
2 (1.12%)2 (1.57%)0 (0.00%)
Grade 0.4221.570.58–4.04
-
Low (G1/G2)
148 (82.68%)106 (83.46%)42 (80.77%)
-
High (G3)
26 (14.35%)16 (12.60%)10 (19.23%)
-
Unknown
5 (2.79%)5 (3.94%)0 (0.00%)
LVSI 0.008 *2.521.24–5.21
-
Positive
77 (43.02%)46 (36.22%)31 (59.62%)
-
Negative
100 (55.87%)79 (62.20%)21 (40.38%)
-
Unknown
2 (1.12%)2 (1.57%)0 (0.00%)
Estrogen receptors 0.7652.250.24–109.25
-
Positive
135 (75.42%)93 (73.23%)42 (80.77%)
-
Negative
6 (3.35%)5 (3.94%)1 (1.92%)
-
Unknown
38 (21.23%)29 (22.83%)9 (17.31%)
Progesterone receptors 0.3523.710.47–169.42
-
Positive
132 (73.74%)90 (70.87%)42 (80.77%)
-
Negative
9 (5.03%)8 (6.30%)1 (1.92%)
-
Unknown
38 (21.23%)29 (22.83%)9 (17.31%)
P53 0.3350.500.11–1.64
-
Aberrant
22 (12.29%)18 (14.17%)4 (7.69%)
-
Wild-type
156 (87.15%)108 (85.04%)48 (92.31%)
-
Unknown
1 (0.56%)1 (0.79%)0 (0.00%)
Myometrial invasion 0.9021.100.54–2.28
-
≥50%
104 (58.10%)73 (57.48%)31 (59.62%)
-
<50%)
72 (40.22%)52 (40.94%)20 (38.46%)
-
Unknown
3 (1.68%)2 (1.57%)1 (1.92%)
* To highlight statistically significant values.
Table 4. Multivariable logistic regression with Wald tests—outcome: dMMR vs. pMMR. AUC = [0.646], 95% CI [0.546–0.735]. Complete cases = 169; events (dMMR) = 51. OR—odds ratio; CI—confidence interval.
Table 4. Multivariable logistic regression with Wald tests—outcome: dMMR vs. pMMR. AUC = [0.646], 95% CI [0.546–0.735]. Complete cases = 169; events (dMMR) = 51. OR—odds ratio; CI—confidence interval.
VariableOR95% CIp-Value
FIGO (III/IV)1.950.72–5.250.187
Grade (high)1.260.48–3.280.642
LVSI (present)2.961.25–7.040.014 *
P53 (aberrant)0.880.25–3.030.838
Myometrial invasion (deep)0.500.21–1.210.123
* To highlight statistically significant values.
Table 5. Multivariable logistic regression with Wald tests—outcome: LVSI present vs. absent. AUC = [0.807], 95% CI [0.743–0.867]. Complete cases = 169; events (LVSI present) = 72. OR—odds ratio; CI—confidence interval.
Table 5. Multivariable logistic regression with Wald tests—outcome: LVSI present vs. absent. AUC = [0.807], 95% CI [0.743–0.867]. Complete cases = 169; events (LVSI present) = 72. OR—odds ratio; CI—confidence interval.
VariableOR95% CIp-Value
MMR (dMMR)3.381.46–7.810.004 *
Grade (high)1.740.64–4.750.281
P53 (aberrant)0.530.16–1.720.289
Myometrial invasion (deep)12.455.15–30.11<0.001 *
* To highlight statistically significant values.
Table 6. Multivariable logistic regression with Wald tests—outcome: FIGO I/II vs. III/IV. AUC = [0.709], 95% CI [0.604–0.811]. Complete cases = 173; events (FIGO III/IV) = 26. OR—odds ratio; CI—confidence interval.
Table 6. Multivariable logistic regression with Wald tests—outcome: FIGO I/II vs. III/IV. AUC = [0.709], 95% CI [0.604–0.811]. Complete cases = 173; events (FIGO III/IV) = 26. OR—odds ratio; CI—confidence interval.
VariableOR95% CIp-Value
MMR (dMMR)2.681.11–6.470.028 *
Grade (high)3.851.45–10.190.007 *
P53 (aberrant)1.370.35–5.370.654
* To highlight statistically significant values.
Table 7. Combinations of loss of expression of MMR-related proteins.
Table 7. Combinations of loss of expression of MMR-related proteins.
Pattern of MMR
Protein Loss
Number of Cases
(Percent)
Pattern of MMR
Protein Loss
Number of Cases
(Percent)
MLH1, PMS241 (78.8%)MLH1, PMS2, MSH61 (1.9%)
MSH64 (7.7%)MSH21 (1.9%)
MLH12 (3.8%)PMS2, MSH2, MSH61 (1.9%)
MSH2, MSH62 (3.8%)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Miedziarek, C.; Bochyński, H.; Bociańska, K.; Potograbski, M.; Tyburski, P.; Zaborowski, M.P.; Nowak-Markwitz, E. Association Between MMR Status and Prognostic Pathological Factors in Endometrioid Endometrial Cancer—A Single-Center Retrospective Study. Cancers 2025, 17, 3605. https://doi.org/10.3390/cancers17223605

AMA Style

Miedziarek C, Bochyński H, Bociańska K, Potograbski M, Tyburski P, Zaborowski MP, Nowak-Markwitz E. Association Between MMR Status and Prognostic Pathological Factors in Endometrioid Endometrial Cancer—A Single-Center Retrospective Study. Cancers. 2025; 17(22):3605. https://doi.org/10.3390/cancers17223605

Chicago/Turabian Style

Miedziarek, Cezary, Hubert Bochyński, Katarzyna Bociańska, Michał Potograbski, Piotr Tyburski, Mikołaj Piotr Zaborowski, and Ewa Nowak-Markwitz. 2025. "Association Between MMR Status and Prognostic Pathological Factors in Endometrioid Endometrial Cancer—A Single-Center Retrospective Study" Cancers 17, no. 22: 3605. https://doi.org/10.3390/cancers17223605

APA Style

Miedziarek, C., Bochyński, H., Bociańska, K., Potograbski, M., Tyburski, P., Zaborowski, M. P., & Nowak-Markwitz, E. (2025). Association Between MMR Status and Prognostic Pathological Factors in Endometrioid Endometrial Cancer—A Single-Center Retrospective Study. Cancers, 17(22), 3605. https://doi.org/10.3390/cancers17223605

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop