Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Exploring biomarkers and prognostic factors in uterine carcinosarcoma: An insight into L1CAM, CDX2, p53, and MSI status

  • Jesse Lopes da Silva ,

    Roles Conceptualization, Data curation, Investigation, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing

    jesse.silva@inca.gov.br

    Affiliation Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Lucas Zanetti de Albuquerque,

    Roles Formal analysis, Investigation, Validation, Writing – original draft, Writing – review & editing

    Affiliation Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Fabiana Resende Rodrigues,

    Roles Formal analysis, Methodology, Validation, Writing – original draft, Writing – review & editing

    Affiliation Division of Pathology, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Nina Carrossini Bastos,

    Roles Conceptualization, Methodology, Project administration, Validation, Writing – review & editing

    Affiliation Division of Pathology, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Isabele Avila Small,

    Roles Conceptualization, Formal analysis, Investigation, Writing – review & editing

    Affiliation Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Elisa Bouret Campos Barroso,

    Roles Investigation, Resources, Validation, Writing – original draft

    Affiliation Clinical Oncology Section, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Fernando Lopes Cordero,

    Roles Conceptualization, Investigation, Methodology, Writing – original draft

    Affiliation Gynecologic Oncology Section, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Daniel de Souza Fernandes,

    Roles Conceptualization, Data curation, Resources, Visualization, Writing – original draft

    Affiliation Gynecologic Oncology Section, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Eduardo Paulino,

    Roles Conceptualization, Investigation, Resources, Writing – original draft

    Affiliation Clinical Oncology Section, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

  • Andreia Cristina de Melo

    Roles Conceptualization, Data curation, Methodology, Validation, Writing – original draft, Writing – review & editing

    Affiliation Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil

Abstract

Background

Uterine Carcinosarcomas (UCS) are a rare type of cancer composed of an admixture of high-grade carcinomatous and sarcomatous elements. Clinicopathological prognostic factors in UCS are well established, but studies that approach the impact of biomarkers in this unusual disease are scarce. The study objective was to evaluate the prevalence and prognostic impact of a panel of prominent biomarkers in uterine carcinosarcoma (UCS) using an immunohistochemical characterization with four biomarkers.

Methods and findings

The internal database of a single Brazilian institution was carefully explored to select women diagnosed with UCS who were submitted to surgery and postoperative chemotherapy with carboplatin and paclitaxel between January 2012 and December 2017. Tissue microarrays containing UCS samples were evaluated by immunohistochemistry for L1CAM, CDX2, p53 and microsatellite instability markers. A total of 57 cases were included. The mean age was 65.3 years (standard deviation, SD 7.0). L1CAM was negative (score 0, no staining) in 27 (47.4%) patients. Of L1CAM-positive, 10 (17.5%) showed weak (score 1, <10%), 6 (10.5%) showed moderate (score 2, between 10–50%), and 14 (24.6%) showed strong L1CAM staining (score 3, ≧50%). dMMR occurred in 3 (5.3%) cases. The p53 was aberrantly expressed in 15 (26.3%) tumors. CDX2 was positive in 3 (5.3%) patients. The three-year progression-free survival (PFS) rate in the general population of the study was 21.2% (95% CI: 11.7–38.1) and the three-year overall survival (OS) rate was 29.4% (95% CI: 18.1–47.6). By multivariate analysis, the presence of metastases and CDX2-positive were significantly associated with poorer PFS (p < 0.001 and p = 0.002, respectively) and OS (p < 0.001 and p = 0.009, respectively).

Conclusion

The strong influence of CDX2 on prognosis requires further investigation. Biological or molecular variability may have impaired the assessment of the impact of the other markers on survival.

Introduction

Uterine Carcinosarcomas (UCS) are a rare type of gynecological tumors that represent less than 5% of endometrial cancers, usually affecting postmenopausal patients and presenting with aggressive behavior [1]. Unlike endometrioid tumors, 45% of cases are diagnosed as FIGO stage III or IV [2, 3]. They are composed of an admixture of high-grade carcinomatous and sarcomatous elements [4, 5]. It is now accepted that UCS are a subtype of epithelial endometrial carcinomas, where the sarcoma is derived from the carcinoma as a result of transdifferentiation during tumor evolution in a process called epithelial-mesenchymal transition (EMT) [6]. Together with the fact that most metastases contain the epithelial counterpart (as high as 90%) [7], platinum doublets are the regimen of choice for adjuvant and palliative settings. Known prognostic factors in UCS include clinical stage, tumors bigger than 5 cm, deep myometrial invasion, lymphovascular space invasion and sarcoma predominance [2, 79].

Initially identified in neuronal cells involved in axon outgrowth and guidance during central nervous system development, the L1-cell adhesion molecule (L1CAM) is a transmembrane molecule of 200- to 220-kDa critical for embryonic development [10]. Some of the main pathways responsible for the EMT process involve the downregulation of E-Cadherin and hormonal receptors and the upregulation of L1CAM [11]. This adhesion molecule is highly expressed in ovarian and endometrial carcinomas and correlates with advanced-stage disease, metastasis and shorter survival [12]. In vitro studies suggest that the cleavage and release of soluble particles of L1CAM in serum may participate in the processes of migration, invasion and escape from programmed cell death [13, 14].

The Caudal-related homeobox transcription factor 2 (CDX2) is frequently expressed in both neoplastic and non-neoplastic cells within the gastrointestinal system [15]. The diffuse and intense immunohistochemical expression of CDX2 is useful in distinguishing colorectal adenocarcinoma from other sites [16, 17]. Recent reports have also identified the presence of this marker in other sites, such as ovarian epithelial tumors (especially in mucinous tumors), pancreatic adenocarcinoma, and transitional cell bladder carcinoma [1821]. However, some small reports have reported varying rates of CDX2 expression in endometrioid adenocarcinoma (1.4–27%) [20, 2224]. While it is well-established that colorectal tumors with CDX2 expression have better overall and cancer-specific survival rates, the prognostic impact of CDX2 on gynecological tumors, including UCS, is underexplored [25].

By inactivating the p53 protein (p53), which is an essential factor for controlling cell growth, some mutations of the TP53 suppressor gene are likely to play a direct role in carcinogenesis [26]. Mutant p53, which has a longer half-life compared to the wild-type protein, can be detected through immunohistochemical assays and has been identified as a marker of poorer outcomes for endometrial tumors [27]. Nonetheless, data on p53 expression in carcinosarcoma is limited [28].

Microsatellite Instability (MSI) is a crucial pathway in the development of endometrial cancer and has emerged as a significant predictor of response to immunotherapy and survival in tumors from various sites, including endometrial adenocarcinomas [2931]. The loss of expression of mismatch repair (MMR) enzymes MLH1, PMS2, MSH2, and MSH6, which can be detected using immunohistochemistry (IHC), may serve as a surrogate marker for expensive and inaccessible genetic testing, which remains the gold standard for detecting cases with high-MSI [29, 32]. However, the data on endometrial carcinosarcoma is still insufficient.

The identification of accessible biomarkers with clinical utility for the management of UCS still poses an unmet need. The current study aimed to provide a description of the prevalence and explore the prognostic value of L1CAM, CDX2, p53 and microsatellite instability status in patients with UCS.

Materials & methods

Ethics statement, study design, patient selection and data collection

This study has been approved by the Ethics in Human Research Committee of the Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil, and was conducted following Good Clinical Practice guidelines [33]. As this is an observational retrospective study, the Institutional Review Board decided to waive the requirement for informed consent from all patients. The authors strictly followed the recommendations of the STrengthening the Reporting of OBservational studies in Epidemiology (STROBE) guidelines and were involved in the study design, writing, critical review and editing of the manuscript, and vouched for the accuracy and completeness of the reported data.

All women diagnosed with UCS who underwent surgery and received postoperative chemotherapy with the standard dose of carboplatin and paclitaxel every three weeks for six cycles [34] at INCA between January 2012 and December 2017 were identified through a meticulous search in the institutional database. Patients with inadequate or insufficient pathological samples and synchronous or anachronistic tumors were excluded from this cohort. Clinical data, including sociodemographic factors, staging, surgery, histological subtype (homologous versus heterologous), disease progression and survival were retrospectively obtained from medical records. The staging was performed based on the International Federation of Gynecology and Obstetrics (FIGO) criteria of 2009 [35].

Immunohistochemistry

The tissue microarray (TMA) assembly was constructed from regions of the formalin-fixed paraffin-embedded surgical specimen of primary tumors with the highest tumor cellularity. The sample cores were punched at six 4-μm intervals, and sarcomatous and epithelial areas were well-represented. Negative controls were created by counterstaining with hematoxylin, while positive controls were used for comparison with the tumor cell staining. Additionally, the slides were evaluated based on the percentage of positive cells compared to the total number of cells.

The expression of L1CAM (Clone 14.10, Biolegend, diluted at 1:1000) in tumor cells was evaluated based on the degree of positivity, with a score of 0 indicating 0% of positivity, a score of 1 indicating 1–10%, a score of 2 indicating ≥10–50%, and a score of 3 indicating ≥50%. For survival analysis, L1CAM positivity was defined as ≥1% (score ≥1) of tumor cells showing membranous L1CAM staining [36]. CDX2 expression (clone EPR2764Y, Cell Marque, diluted at 1:700) was considered positive if the score was ≥1% [24]. Aberrant p53 expression (clone SP53, Cell Marque, diluted at 1:500) was defined as ≥80% of positive tumor cell nuclei [37]. Loss of expression was determined if there was no nuclear staining for any of MLH1 (clone G168-728, Cell Marque, diluted at 1:600), PMS2 (clone MRQ-28, Cell Marque, diluted at 1:700), MSH2 (clone G219-1129, Cell Marque, diluted at 1:1000), and MSH6 (clone 44, Cell Marque, diluted at 1:300). The IHC status was then classified into two groups: mismatch repair deficient (dMMR) if at least one marker lacked nuclear staining and mismatch repair proficient (pMMR) if all markers showed nuclear staining [38].

Statistical analysis

Progression-free survival (PFS) was determined by measuring the time from the first administration of CP infusion until the earliest date of disease progression, recurrence, or death. Overall survival (OS) was measured from the initial CP infusion until death from any cause or until the patient’s status was censored if they were alive on the last day of data collection. The Kaplan-Meier method was utilized to estimate the median duration of PFS and OS. Patients were categorized based on an array of factors such as their age, body mass index (BMI), race, stage, omentectomy, residual disease, adjuvant radiotherapy, the existence of lymphovascular invasion (LVI), histological subtype, and the status of IHC markers. The Shapiro-Wilk test was used to evaluate the normality of continuous variables, while categorical variables were presented as absolute and relative frequencies.

To evaluate the association of IHC findings and clinicopathological features with staging, the Student’s t-test or χ2 test for p-values were employed. The Cox proportional hazards were used to calculate the crude Hazard Ratio (HR) for each variable. For multivariate analysis, all variables with an association with survival outcomes at a p-value < 0.30 were included, and the Akaike Information Criterion (AIC) was employed to select the most appropriate model for multiple Cox analysis. A p-value < 0.05 was considered statistically significant, and any missing data were excluded from the analysis. The R project version 3.5.3 was used to conduct the statistical analyses [39].

Results

In this cohort, 57 women were examined. At the time of diagnosis, the average age was 65.3 years (range: 49 to 79 years). The majority of patients were non-Caucasian (42 cases, which accounts for 73.7% of the cohort), and most had advanced disease (42 cases, or 73.7%, had FIGO III/IV). Additionally, the heterologous subtype was present in 30 cases (52.6%) and positive LVI was found in 25 cases (43.9%).

Regarding the initial surgical approach, 37 patients (65%) underwent lymphadenectomy, 27 patients (47.4%) underwent omentectomy, and 39 patients (68.4%) had an optimal resection, which was defined as having a residual disease size of less than 1.0 cm. All patients received postoperative chemotherapy with carboplatin plus paclitaxel as per the inclusion criteria. Adjuvant radiotherapy was subsequently performed in 24 patients (42.8%). The clinicopathological characteristics are summarized in Table 1, while Table 2 provides information on the surgical and adjuvant approaches.

thumbnail
Table 1. Correlation of clinical-pathological characteristics and clinical stage.

https://doi.org/10.1371/journal.pone.0285447.t001

thumbnail
Table 2. Treatment data of the study population (N = 57).

https://doi.org/10.1371/journal.pone.0285447.t002

As shown in Table 1, L1CAM was completely negative (score 0) in 27 (47.4%) patients. In L1CAM-positive tumors, 10 (17.5%) showed weak expression (score 1), 6 (10.5%) showed moderate (score 2), and 14 (24.6%) showed strong L1CAM staining (score 3). dMMR occurred in 3 (5.3%) cases. For p53, 15 (26.3%) tumors were aberrant. Finally, CDX2 was positive in 3 (5.3%) patients. There was no correlation between clinicopathological characteristics and IHQ expression. Fig 1 shows representative images of cases with high expression of p53, CDX2, MLH1, MSH2, MSH6 and PMS2 markers. Fig 2 displays the graduation score for L1CAM staining.

thumbnail
Fig 1. Immunohistochemistry biomarkers staining representative with high expression of p53, CDX2, MSH2, MLH1, MSH6 and PMS2.

The visual data were captured at a magnification of x20 to ensure precision and are presented in this figure.

https://doi.org/10.1371/journal.pone.0285447.g001

thumbnail
Fig 2. Scoring the L1CAM expression level in graduation ranks.

The L1CAM expression level was assessed and scored as 0, 1+, 2+, and 3+ (A, B, C, and D, respectively) to quantify its intensity. The visual data were captured at a magnification of x20 to ensure precision and are presented in this figure.

https://doi.org/10.1371/journal.pone.0285447.g002

The median follow-up was 51 months (95% confidence interval, CI: 40–70), during which 42 patients experienced disease progression or died. The median PFS was 15 months (95% CI: 10–27), with a three-year PFS rate of 21.2% (95% CI: 11.7–38.1). Univariate analysis revealed that patients with FIGO stages III/IV (HR 2.7; 95% CI: 1.19–6.13; p = 0.017), metastasis (HR 3.28; 95% CI: 1.66–6.49; p = 0.001), residual disease (HR 4.19; 95% CI: 2.13–8.25; p < 0.001), and CDX2-positive tumors (HR 4.16; 95% CI: 1.21–14.32; p = 0.024) had a significantly higher risk of PFS events. For multivariate analysis of PFS, the Cox model included three variables: metastasis (present versus absent; HR 4.06; 95% CI: 1.98–8.34; p < 0.001), L1CAM (positive versus negative HR 0.65; 95% CI: 0.35–1.19; p = 0.162), and CDX2 (positive versus negative; HR 7.81; 95% CI: 2.10–28.99; p = 0.002). Table 3 provides a summary of the univariate and multivariate analyses for PFS.

thumbnail
Table 3. Crude and adjusted hazards ratios for carcinosarcoma progression-free survival (PFS) estimated by univariate analysis and multivariate analysis.

https://doi.org/10.1371/journal.pone.0285447.t003

At the time of analysis, 38 patients had died. The median OS was 28 months (95% CI: 15–53), with a three-year OS rate of 29.4% (95% CI: 18.1–47.6). Univariate analysis demonstrated that patients with FIGO stage III/IV (HR 3.62; 95% CI: 1.40–9.35; p = 0.008), metastasis (HR 3.23; 95% CI: 1.61–6.46; p = 0.001), residual disease (HR 3.83; 95% CI: 1.92–7.62); p < 0.001), and CDX2-positive tumors (HR 3.42; 95% CI: 1.01–11.60; p = 0.048) had a significantly higher risk of death. For multivariate analysis of OS, the Cox model also included three variables: metastasis (present versus absent; HR 3.93; 95% CI: 1.90–8.15; p < 0.001), L1CAM (positive versus negative; HR 0.57; 95% CI: 0.29–1.09; p = 0.091), and CDX2 (positive versus negative; HR 5.50; 95% CI: 1.54–19.69; p = 0.009). Table 4 provides a summary of the univariate and multivariate analyses for OS. Figs 3 and 4 depict the Kaplan-Meier curves for PFS and OS, respectively, according to the analyzed variables.

thumbnail
Fig 3. Progression-free survival (PFS).

(A) stage status; (B) residual disease status (C) CDX2 status. Stratification of residual disease after surgery considered R0 when there was no evidence of residual disease and R1 or R2 correlate to microscopic and macroscopic residual disease, respectively. For immunohistochemistry biomarkers, Kaplan Meier curves for PFS were stratified by the median values as the cut-off for prognostic evaluation and divided into low vs high molecule expression patterns. The blue solid line indicates patients with high values and the red solid line low values. Tick marks indicate censored data.

https://doi.org/10.1371/journal.pone.0285447.g003

thumbnail
Fig 4. Overall survival (OS).

(A) stage status (B) residual disease status (C) CDX2 status. Stratification of residual disease after surgery considered R0 when there was no evidence of residual disease and R1 or R2 correlate to microscopic and macroscopic residual disease, respectively. For immunohistochemistry biomarkers, Kaplan Meier curves for PFS were stratified by the median values as the cut-off for prognostic evaluation and divided into low vs high molecule expression patterns. The blue solid line indicates patients with high values and the red solid line low values. Tick marks indicate censored data.

https://doi.org/10.1371/journal.pone.0285447.g004

thumbnail
Table 4. Crude and adjusted hazards ratios for carcinosarcoma overall survival (OS) estimated by univariate analysis and multivariate analysis.

https://doi.org/10.1371/journal.pone.0285447.t004

Discussion

UCS is a rare disease, characterized by more advanced stages at diagnosis and poorer prognosis when compared to endometrioid carcinomas. The treatment generally consists of primary surgery followed by adjuvant chemoradiotherapy. In advanced disease, surgery can be considered the first approach when complete cytoreduction is deemed feasible. Regarding systemic therapy, carboplatin plus paclitaxel is the preferred regimen since it is likely to be non-inferior to paclitaxel and ifosfamide based on the results of the recently published GOG-0261 trial [40]. By the time the women included in the study were treated, platinum doublets had already been incorporated into the INCA routine as the standard of care for UCS.

The clinicopathological and sociodemographic characteristics observed in the current cohort are consistent with those reported by Matsuo et al. in a larger national database [1, 2, 41]. Of particular note are the higher prevalence of women over 60 years of age, obese individuals, and those with heterologous subtype tumors, as well as a significant presence of LVI. The proportion of cases diagnosed at advanced stages was considerable, with some patients having metastases detected only during or after surgery. The rates of omentectomy and lymphadenectomy in our cohort were also consistent with previously reported series [4244].

As expected, the current analysis revealed a significant association between advanced tumor stage, metastasis, and residual disease with shorter PFS and OS. Possibly, the exclusion of staging and residual disease variables from the selected model based on the Akaike criteria was likely due to the strong correlation between these two variables impairing the statistical analysis, although no specific test was performed to confirm this hypothesis. Previous studies have already demonstrated the favorable impact of complete cytoreduction of UCS on OS. For instance, Tanner et al. reported a more favorable outcome in patients who achieved complete cytoreduction compared to those who underwent suboptimal surgery in a cohort of 44 patients with advanced carcinosarcoma (median OS of 52 versus 9 months, respectively, p < 0.0001) [45].

The mutational profile of UCS has not been thoroughly examined so far, unlike the endometrioid carcinoma setting. Although CDX2 was not previously linked to poorer outcomes in UCS, the current analysis reveals that it is infrequent in this population (5.3% of cases) but with a significant impact on prognosis. This finding may suggest that CDX2 is likely to be a marker of more aggressive behavior in UCS. CDX2 is highly expressed in various gastrointestinal cancers, such as colorectal adenocarcinomas (70–85%), gastroesophageal (50%), stomach (55–70%), ampulla (65–80%) and small intestine tumors (60%) [20, 46]. It can also be found in some non-gastrointestinal cancers, such as urachal, pulmonary mucinous, and certain gynecological tumors. In their study, Park et al. reported an incidence rate of 3.9% for malignant mixed Müllerian tumor cases [22]. In contrast, the other IHC markers (L1CAM, p53, and dMMR) used in this cohort did not significantly influence survival.

The Cancer Genome Atlas (TCGA) Research Network has identified four molecular subtypes of endometrioid and serous endometrial cancer based on the mutational burden, levels of copy number alterations, and MSI status. These subtypes include the POLE ultramutated group, the MSI hypermutated group, the copy number-high (serous-like) group, and the copy number-low (endometrioid) group, each of which has been associated with varying prognosis and progression-free survival outcomes [4749]. In uterine carcinosarcomas (UCS), the majority of cases (60–78%) are classified as copy-number-high, with a smaller proportion of cases (22–38%) classified as copy-number-low. Only a minority of endometrial carcinosarcomas (< 5%) belong to the ultramutated or hypermutated groups [6, 47, 50]. The prevalence of p53 overexpression in UCS has been reported to range from 60–90% in previous studies, which is higher than the rates observed in the current cohort [6, 5155]. However, methodological differences may explain this discrepancy. For both endometrioid and serous endometrial tumors, p53 overexpression is strongly associated with worse outcomes. In one study, Segura et al. found similar rates of p53 overexpression (20%) and dMMR (6.2%) in their cohort and described better outcomes for those UCS with dMMR compared to traditional pMMR [56]. In another study, Gotoh et al. found significant correlations between POLE, MSI, CNH, and CNL subtypes with patient outcomes in terms of progression-free survival and overall survival [6]. While the current study was not able to perform gene sequencing, the results for p53 and dMMR in this cohort were opposite to those reported by Gotoh et al. and Segura et al.

In lower-risk histologies, specifically endometrioid tumors, L1CAM has impactful prognostic value [10, 36, 57, 58]. The expression of p53 and L1CAM is positively correlated, with around 60–70% of p53 aberrant tumors also exhibiting L1CAM positivity (10% or higher) [10, 36, 57, 58]. However, in this population, L1CAM has not been shown to provide additional prognostic information [10, 36, 57, 58]. In the UCS scenario, the lack of prognostic impact of L1CAM may be explained by the increased expression of L1CAM that occurs only after the epithelial-mesenchymal transition (EMT) induction phase through exposure to the cytokine TGF-B1, which results in the upregulation of the EMT transcription factor Slug [58]. It is theoretically possible that cell migration and invasion would be more intensively triggered if this L1CAM upregulation process occurred at earlier stages [59]. The most significant contribution of L1CAM expression in tumor development may occur during the early stage of EMT, when cancer cells gain motility and invasive properties. This process could occur through direct action as an adhesion molecule or indirectly by acting as a cell-signaling molecule [11]. However, at a later stage of carcinogenesis, as may be the case for most patients in the present study, motility and invasive properties may have less impact on tumor progression and the metastasis process [60].

The strengths of this research rely primarily on the originality of the comprehensive evaluation of potentially valuable accessible biomarkers in the management of uterine UCS. The strict inclusion criteria ensured the selection of patients with homogeneous characteristics, favoring the assessment of the impact of clinicopathological features on disease progression and survival. The surgical samples were blindly examined by two experienced pathologists, and the results were consolidated using multivariate analyzes to enhance internal validity.

Conversely, the limitations of this study are closely related to its retrospective nature. In consequence, some confounding factors may be missing, which could weaken the quality of the analysis. Molecular analysis was not performed in this study, and opportunities to offer multimodal adjuvant treatment with chemotherapy followed by radiotherapy were decided by the care providers. Lastly, the small sample size may have been inadequate to ensure adequate statistical power to detect slight differences in survival for some IHC markers.

Conclusion

Given the infrequency of UCS, conducting prospective randomized studies can be challenging. The current study assesses a panel of four biomarkers and presents some unprecedented findings in the field of UCS. The noteworthy impact of CDX2 on prognosis observed in this cohort warrants further investigation through multicenter studies with more extensive sample sizes. The survival outcomes were not influenced by the status of the other markers, implying the possibility of underlying biological or molecular variations.

Acknowledgments

The authors express their profound gratitude to all the patients and their families who, through their invaluable trust and participation, made this study possible. The authors also acknowledge the indispensable contribution of the provision of biological material for research purposes, which was instrumental in advancing our understanding of this complex disease. Without the selflessness and generosity of patients and their families, this study could not have been accomplished. The authors are humbled and honored to have had the opportunity to conduct this research and to contribute to the advancement of knowledge in this field.

References

  1. 1. Matsuo K, Ross MS, Machida H, Blake EA, Roman LD. Trends of uterine carcinosarcoma in the United States. J Gynecol Oncol. 2018 Mar;29(2):e22. pmid:29400015
  2. 2. Matsuo K, Takazawa Y, Ross MS, Elishaev E, Podzielinski I, Yunokawa M, et al. Significance of histologic pattern of carcinoma and sarcoma components on survival outcomes of uterine carcinosarcoma. Ann Oncol. 2016 Jul 1;27(7):1257–66. pmid:27052653
  3. 3. Abdulfatah E, Lordello L, Khurram M, Van de Vijver K, Alosh B, Bandyopadhyay S, et al. Predictive Histologic Factors in Carcinosarcomas of the Uterus: A Multi-institutional Study. Int J Gynecol Pathol Off J Int Soc Gynecol Pathol. 2019 May;38(3):205–15. pmid:30958427
  4. 4. Cantrell LA, Blank SV, Duska LR. Uterine carcinosarcoma: A review of the literature. Gynecol Oncol. 2015 Jun;137(3):581–8. pmid:25805398
  5. 5. Gorai I, Yanagibashi T, Taki A, Udagawa K, Miyagi E, Nakazawa T, et al. Uterine carcinosarcoma is derived from a single stem cell: an in vitro study. Int J Cancer. 1997 Sep 4;72(5):821–7. pmid:9311600
  6. 6. Gotoh O, Sugiyama Y, Takazawa Y, Kato K, Tanaka N, Omatsu K, et al. Clinically relevant molecular subtypes and genomic alteration-independent differentiation in gynecologic carcinosarcoma. Nat Commun. 2019 Oct 31;10(1):4965. pmid:31672974
  7. 7. Ferguson SE, Tornos C, Hummer A, Barakat RR, Soslow RA. Prognostic features of surgical stage I uterine carcinosarcoma. Am J Surg Pathol. 2007 Nov;31(11):1653–61. pmid:18059221
  8. 8. Gonzalez Bosquet J, Terstriep SA, Cliby WA, Brown-Jones M, Kaur JS, Podratz KC, et al. The impact of multi-modal therapy on survival for uterine carcinosarcomas. Gynecol Oncol. 2010 Mar;116(3):419–23. pmid:19896181
  9. 9. Kurnit KC, Previs RA, Soliman PT, Westin SN, Klopp AH, Fellman BM, et al. Prognostic factors impacting survival in early stage uterine carcinosarcoma. Gynecol Oncol. 2019 Jan;152(1):31–7. pmid:30414738
  10. 10. Versluis M, Plat A, de Bruyn M, Matias-Guiu X, Trovic J, Krakstad C, et al. L1CAM expression in uterine carcinosarcoma is limited to the epithelial component and may be involved in epithelial–mesenchymal transition. Virchows Arch. 2018 Nov 1;473(5):591–8. pmid:30140948
  11. 11. Huszar M, Pfeifer M, Schirmer U, Kiefel H, Konecny GE, Ben-Arie A, et al. Up-regulation of L1CAM is linked to loss of hormone receptors and E-cadherin in aggressive subtypes of endometrial carcinomas. J Pathol. 2010. Apr;220(5):551–61. pmid:20077528
  12. 12. Kommoss FK, Karnezis AN, Kommoss F, Talhouk A, Taran FA, Staebler A, et al. L1CAM further stratifies endometrial carcinoma patients with no specific molecular risk profile. Br J Cance. 2018 Aug;119(4):480–6. pmid:30050154
  13. 13. Thiery JP, Acloque H, Huang RYJ, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009 Nov 25;139(5):871–90. pmid:19945376
  14. 14. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 2011 Mar 25;331(6024):1559–64. pmid:21436443
  15. 15. Suh E, Chen L, Taylor J, Traber PG. A homeodomain protein related to caudal regulates intestine-specific gene transcription. Mol Cell Biol. 1994 Nov;14(11):7340–51. pmid:7935448
  16. 16. Groisman GM, Meir A, Sabo E. The value of Cdx2 immunostaining in differentiating primary ovarian carcinomas from colonic carcinomas metastatic to the ovaries. Int J Gynecol Pathol Off J Int Soc Gynecol Pathol. 2004 Jan;23(1):52–7. pmid:14668551
  17. 17. Baba Y, Nosho K, Shima K, Freed E, Irahara N, Philips J, et al. Relationship of CDX2 Loss with Molecular Features and Prognosis in Colorectal Cancer: Implications for Clinical and Pathology Practice. Clin Cancer Res Off J Am Assoc Cancer Res. 2009 Jul 15;15(14):4665–73.
  18. 18. Barbareschi M, Murer B, Colby TV, Chilosi M, Macri E, Loda M, et al. CDX-2 homeobox gene expression is a reliable marker of colorectal adenocarcinoma metastases to the lungs. Am J Surg Pathol. 2003 Feb;27(2):141–9. pmid:12548159
  19. 19. Kaimaktchiev V, Terracciano L, Tornillo L, Spichtin H, Stoios D, Bundi M, et al. The homeobox intestinal differentiation factor CDX2 is selectively expressed in gastrointestinal adenocarcinomas. Mod Pathol Off J U S Can Acad Pathol Inc. 2004 Nov;17(11):1392–9. pmid:15205684
  20. 20. De Lott LB, Morrison C, Suster S, Cohn DE, Frankel WL. CDX2 is a useful marker of intestinal-type differentiation: a tissue microarray-based study of 629 tumors from various sites. Arch Pathol Lab Med. 2005 Sep;129(9):1100–5. pmid:16119980
  21. 21. Moskaluk CA, Zhang H, Powell SM, Cerilli LA, Hampton GM, Frierson HF. Cdx2 protein expression in normal and malignant human tissues: an immunohistochemical survey using tissue microarrays. Mod Pathol Off J U S Can Acad Pathol Inc. 2003 Sep;16(9):913–9. pmid:13679455
  22. 22. Park KJ, Bramlage MP, Ellenson LH, Pirog EC. Immunoprofile of adenocarcinomas of the endometrium, endocervix, and ovary with mucinous differentiation. Appl Immunohistochem Mol Morphol AIMM. 2009 Jan;17(1):8–11. pmid:18776815
  23. 23. Huang LP, Yu YH, Sheng C, Wang SH. Up-regulation of cadherin 17 and down-regulation of homeodomain protein CDX2 correlate with tumor progression and unfavorable prognosis in epithelial ovarian cancer. Int J Gynecol Cancer Off J Int Gynecol Cancer Soc. 2012 Sep;22(7):1170–6. pmid:22810971
  24. 24. Arık DA, Kabukçuoğlu SK. CDX2 expression in endometrial endometrioid adenocarcinoma. Eur J Gynaecol Oncol. 2017 Oct 10;38(5):704–7.
  25. 25. Choi HB, Pyo JS, Son S, Kim K, Kang G. Diagnostic and Prognostic Roles of CDX2 Immunohistochemical Expression in Colorectal Cancers. Diagn Basel Switz. 2022 Mar 20;12(3):757. pmid:35328309
  26. 26. Stelloo E, Nout RA, Osse EM, Jürgenliemk-Schulz IJ, Jobsen JJ, Lutgens LC, et al. Improved Risk Assessment by Integrating Molecular and Clinicopathological Factors in Early-stage Endometrial Cancer-Combined Analysis of the PORTEC Cohorts. Clin Cancer Res Off J Am Assoc Cancer Res. 2016 Aug 15;22(16):4215–24. pmid:27006490
  27. 27. Fadare O, Gwin K, Desouki MM, Crispens MA, Jones HW, Khabele D, et al. The clinicopathologic significance of p53 and BAF-250a (ARID1A) expression in clear cell carcinoma of the endometrium. Mod Pathol Off J U S Can Acad Pathol Inc. 2013 Aug;26(8):1101–10. pmid:23524907
  28. 28. Costa MJ, Vogelsan J, Young LJ. p53 gene mutation in female genital tract carcinosarcomas (malignant mixed müllerian tumors): a clinicopathologic study of 74 cases. Mod Pathol Off J U S Can Acad Pathol Inc. 1994 Aug;7(6):619–27. pmid:7991520
  29. 29. Hashmi AA, Mudassir G, Hashmi RN, Irfan M, Asif H, Khan EY, et al. Microsatellite Instability in Endometrial Carcinoma by Immunohistochemistry, Association with Clinical and Histopathologic Parameters. Asian Pac J Cancer Prev APJCP. 2019;20(9):2601–6. pmid:31554352
  30. 30. Dunlop MG, Farrington SM, Carothers AD, Wyllie AH, Sharp L, Burn J, et al. Cancer risk associated with germline DNA mismatch repair gene mutations. Hum Mol Genet. 1997 Jan;6(1):105–10. pmid:9002677
  31. 31. Esteller M, Levine R, Baylin SB, Ellenson LH, Herman JG. MLH1 promoter hypermethylation is associated with the microsatellite instability phenotype in sporadic endometrial carcinomas. Oncogene. 1998 Nov 5;17(18):2413–7. pmid:9811473
  32. 32. McMeekin DS, Tritchler DL, Cohn DE, Mutch DG, Lankes HA, Geller MA, et al. Clinicopathologic Significance of Mismatch Repair Defects in Endometrial Cancer: An NRG Oncology/Gynecologic Oncology Group Study. J Clin Oncol. 2016 Sep 1;34(25):3062–8. pmid:27325856
  33. 33. World Medical Association. World Medical Association Declaration of Helsinki: Ethical Principles for Medical Research Involving Human Subjects. JAMA. 2013 Nov 27;310(20):2191–4. pmid:24141714
  34. 34. Powell MA, Filiaci VL, Rose PG, Mannel RS, Hanjani P, Degeest K, et al. Phase II evaluation of paclitaxel and carboplatin in the treatment of carcinosarcoma of the uterus: a Gynecologic Oncology Group study. J Clin Oncol. 2010 Jun 1;28(16):2727–31. pmid:20421537
  35. 35. Creasman W. Revised FIGO staging for carcinoma of the endometrium. Int J Gynaecol Obstet. 2009 May;105(2):109. pmid:19345353
  36. 36. Zeimet AG, Reimer D, Huszar M, Winterhoff B, Puistola U, Azim SA, et al. L1CAM in early-stage type I endometrial cancer: results of a large multicenter evaluation. J Natl Cancer Inst. 2013 Aug 7;105(15):1142–50. pmid:23781004
  37. 37. Okamoto K, Nakamura K, Haraga J, Masuyama H. Aberrant p53 expression in metastatic lymph nodes is a significant predictor of recurrence in stage IIIC endometrial cancer. Research Square [Preprint]. 2022 [posted 2022 Feb 21; cited June 3, 2022].
  38. 38. Luchini C, Bibeau F, Ligtenberg MJL, Singh N, Nottegar A, Bosse T, et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol. 2019 Aug;30(8):1232–43. pmid:31056702
  39. 39. R Core Team (2014). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. http://www.R-project.org/
  40. 40. Powell MA, Filiaci VL, Hensley ML, Huang HQ, Moore KN, Tewari KS, et al. Randomized Phase III Trial of Paclitaxel and Carboplatin Versus Paclitaxel and Ifosfamide in Patients With Carcinosarcoma of the Uterus or Ovary: An NRG Oncology Trial. J Clin Oncol. 2022 Mar 20;40(9):968–77. pmid:35007153
  41. 41. Matsuo K, Takazawa Y, Ross MS, Elishaev E, Yunokawa M, Sheridan TB, et al. Characterizing sarcoma dominance pattern in uterine carcinosarcoma: Homologous versus heterologous element. Surg Oncol. 2018 Sep 1;27(3):433–40. pmid:30217299
  42. 42. Beckmann K, Selva-Nayagam S, Olver I, Miller C, Buckley ES, Powell K, et al. Carcinosarcomas of the Uterus: Prognostic Factors and Impact of Adjuvant Treatment. Cancer Manag Res. 2021 Jun 10;13:4633–45. pmid:34140809
  43. 43. Nemani D, Mitra N, Guo M, Lin L. Assessing the effects of lymphadenectomy and radiation therapy in patients with uterine carcinosarcoma: a SEER analysis. Gynecol Oncol. 2008 Oct;111(1):82–8. pmid:18674808
  44. 44. Park JY, Kim DY, Kim JH, Kim YM, Kim YT, Nam JH. The role of pelvic and/or para-aortic lymphadenectomy in surgical management of apparently early carcinosarcoma of uterus. Ann Surg Oncol. 2010 Mar;17(3):861–8. pmid:19921530
  45. 45. Tanner EJ, Leitao MM, Garg K, Chi DS, Sonoda Y, Gardner GJ, et al. The role of cytoreductive surgery for newly diagnosed advanced-stage uterine carcinosarcoma. Gynecol Oncol. 2011 Dec;123(3):548–52. pmid:21945551
  46. 46. Winn B, Tavares R, Fanion J, Noble L, Gao J, Sabo E, et al. Differentiating the undifferentiated: immunohistochemical profile of medullary carcinoma of the colon with an emphasis on intestinal differentiation. Hum Pathol. 2009 Mar;40(3):398–404. pmid:18992917
  47. 47. Cancer Genome Atlas Research Network, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013 May 2;497(7447):67–73. pmid:23636398
  48. 48. Talhouk A, Hoang LN, McConechy MK, Nakonechny Q, Leo J, Cheng A, et al. Molecular classification of endometrial carcinoma on diagnostic specimens is highly concordant with final hysterectomy: Earlier prognostic information to guide treatment. Gynecol Oncol. 2016 Oct;143(1):46–53. pmid:27421752
  49. 49. León-Castillo A, Gilvazquez E, Nout R, Smit VT, McAlpine JN, McConechy M, et al. Clinicopathological and molecular characterisation of “multiple-classifier” endometrial carcinomas. J Pathol. 2020 Mar;250(3):312–22. pmid:31829447
  50. 50. McConechy MK, Hoang LN, Chui MH, Senz J, Yang W, Rozenberg N, et al. In-depth molecular profiling of the biphasic components of uterine carcinosarcomas. J Pathol Clin Res. 2015 Apr 9;1(3):173–85. pmid:27499902
  51. 51. Cherniack AD, Shen H, Walter V, Stewart C, Murray BA, Bowlby R, et al. Integrated Molecular Characterization of Uterine Carcinosarcoma. Cancer Cell. 2017 Mar 13;31(3):411–23. pmid:28292439
  52. 52. Crane E, Naumann W, Tait D, Higgins R, Herzog T, Brown J. Molecular variations in uterine carcinosarcomas identify therapeutic opportunities. Int J Gynecol Cancer. 2020 Apr;30(4):480–4. pmid:32114514
  53. 53. Jones NL, Xiu J, Chatterjee-Paer S, Buckley de Meritens A, Burke WM, Tergas AI, et al. Distinct molecular landscapes between endometrioid and nonendometrioid uterine carcinomas. Int J Cancer. 2017 Mar 15;140(6):1396–404. pmid:27905110
  54. 54. Szukala SA, Marks JR, Burchette JL, Elbendary AA, Krigman HR. Co-expression of p53 by epithelial and stromal elements in carcinosarcoma of the female genital tract: an immunohistochemical study of 19 cases. Int J Gynecol. 1999 Mar;9(2):131–6. pmid:11240754
  55. 55. Abargel A, Avinoach I, Kravtsov V, Boaz M, Glezerman M, Menczer J. Expression of p27 and p53: comparative analysis of uterine carcinosarcoma and endometrial carcinoma. Int J Gynecol Cancer. 2004 Apr;14(2):354–9. pmid:15086737
  56. 56. Segura S, Pedra Nobre S, Hussein Y, Abu-Rustum N, Weigelt B, Soslow R, et al. DNA mismatch repair-deficient endometrial carcinosarcomas portend distinct clinical, morphological and molecular features compared to traditional carcinosarcomas. Am J Surg Pathol. 2020 Nov;44(11):1573–9. pmid:32804882
  57. 57. Bosse T, Nout RA, Stelloo E, Dreef E, Nijman HW, Jürgenliemk-Schulz IM, et al. L1 cell adhesion molecule is a strong predictor for distant recurrence and overall survival in early stage endometrial cancer: Pooled PORTEC trial results. Eur J Cancer. 2014 Oct 1;50(15):2602–10. pmid:25126672
  58. 58. van der Putten LJ, Visser NC, van de Vijver K, Santacana M, Bronsert P, Bulten J, et al. L1CAM expression in endometrial carcinomas: an ENITEC collaboration study. Br J Cancer. 2016 Sep;115(6):716–24. pmid:27505134
  59. 59. Kiefel H, Bondong S, Pfeifer M, Schirmer U, Erbe-Hoffmann N, Schäfer H, et al. EMT-associated up-regulation of L1CAM provides insights into L1CAM-mediated integrin signalling and NF-κB activation. Carcinogenesis. 2012 Oct 1;33(10):1919–29. pmid:22764136
  60. 60. Kiefel H, Bondong S, Hazin J, Ridinger J, Schirmer U, Riedle S, et al. L1CAM: a major driver for tumor cell invasion and motility. Cell Adhes Migr. 2012 Jul 24;6(4):374–84. pmid:22796939