- Research Note
- Open access
- Published:
Genetic analysis of uterine lavage fluids to identify women at high risk of endometrial cancer
BMC Research Notes volumeĀ 18, ArticleĀ number:Ā 117 (2025)
Abstract
Objectives
Endometrial cancer (EC) is the most common malignancy of the female genital tract in developed countries, yet preventive screening remains unavailable, and diagnostic approaches are largely limited to symptomatic women. Despite advancements in precision oncology, the biology of precancerous lesions is less understood compared to advanced disease. To address this gap, we conducted a prospective case-control study analysing uterine lavage fluid from women undergoing diagnostic evaluation. The study included 257 participants: 80 diagnosed with endometrial intraepithelial neoplasia (EIN), 89 with early-stage EC, and 88 healthy controls. Using targeted next-generation sequencing, we examined genetic alterations in 22 selected genes associated with EC development.
Results
Our findings did not confirm a direct association between specific genetic mutations in uterine lavage fluid and the presence of EIN or early-stage EC (pā=ā0.501). Mutations were detected in both cases and controls, with a higher overall mutation burden observed in controls, suggesting potential background genomic alterations unrelated to EC development. In conclusion, while molecular profiling of uterine lavage fluid remains a promising concept for non-invasive diagnosis, our results highlight significant challenges in specificity. Further studies with larger cohorts and additional biomarkers are necessary to clarify its diagnostic relevance and clinical applicability.
Introduction
Endometrial cancer (EC) is the most prevalent cancer of the female genital tract in developed countries and the sixth most common cancer in women worldwide, with its incidence and mortality steadily rising [1, 2]. Current diagnostic practices rely on the presence of clinical symptoms, meaning that most cases are diagnosed only after symptoms appear, underscoring the lack of effective screening strategies for early detection [3]. Epidemiologically, EC risk is most strongly associated with estrogen exposure, however, other factors such as obesity, diabetes, early menarche, nulliparity, late menopause, advanced age, and tamoxifen use also play significant roles [4, 5]. Clinically, abnormal uterine bleeding or spotting, sometimes accompanied by vaginal discharge, is the most common presenting symptom. The standard diagnostic approach involves ultrasonography followed by dilatation and curettage or curettage followed by hysteroscopy to confirm the diagnosis through histopathological examination.
While targeted prevention efforts are emerging, they are limited by gaps in understanding the biology of precancerous endometrial lesions, as our knowledge largely pertains to advanced disease [6]. Endometrial cancers are broadly classified into two types based on precursor lesions: Type I EC, which commonly originates from atypical hyperplasia, and Type II EC, often arising from atrophic endometrium or within an endometrial polyp. Molecular distinctions between these precancerous lesions and normal endometrial tissue are clear, with Type I EC frequently exhibiting mutations in PTEN, KRAS, and beta-catenin, alongside microsatellite instability [7]. In contrast, Type II EC typically shows TP53 mutations, HER2/neu amplification, and loss of E-cadherin as well as p16 function due to either mutation or hypermethylation [8]. These molecular changes, evident even in early lesions, suggest the potential for early detection through genetic analysis.
In this study, we conducted a prospective analysis of uterine lavage fluid collected from women undergoing diagnostic evaluation, employing targeted next-generation sequencing (NGS) to detect genetic alterations associated with endometrial intraepithelial neoplasia (EIN) and early-stage endometrial carcinoma in comparison with healthy controls. This approach aims to establish a minimally invasive method for identifying high-risk individuals, potentially enabling earlier intervention for EC.
Materials and methods
Patient enrolment and sample collection
The local ethical committee of the University Hospital in Brno (FN Brno) approved the case-control study starting from 1 May 2021 and lasting until 31 December 2024, i.e. 44 months in total, with informed consent obtained from each patient included in the study. Only patients planning to undergo a hysterectomy (due to cancer, precancer, or other reasonsā for example, uterine fibroids, uterine prolapse.) with a final histopathological examination were included in the study. The uterine lavage was done immediately before hysteroscopy or hysterectomy in the operating theatre. Cases were divided into three groups: (a) ECā histologically confirmed endometrial cancer, (b) EINā histologically confirmed endometrial intraepithelial neoplasia, and (c) controlā cases with benign histology. Cases with age under 18 years, with any previous malignity or cancer duplicity as well as with different histology from biopsy and final histological specimen were excluded. Clinical data were evaluated by oncologist from the hospitalās patient records. Besides histological results, potential clinical risk factors for endometrial cancer (age, BMI, parity, hormonal contraceptives, arterial hypertension, and diabetes mellitus) were also recorded.
Fluids from uterine lavages were stored at 4 °C and processed within 24Ā h. Briefly, samples were centrifuged at 3,200 Ćg/ 4 °C for 20Ā min. Pellets were resuspended in erythrocyte buffer (RBC) and incubated for 15Ā min. The samples were then centrifuged again for 20Ā min at 3,200 Ćg/ 4 °C and the resulting pellets were frozen at -80 °C until DNA isolation.
Library preparation and sequencing
DNA was isolated using DNeasy Blood & Tissue Kit (Qiagen, Germany) according to manufacturer“s instructions. Sequencing libraries were prepared using KAPA HyperPlus Library Preparation Kit (Roche, Switzerland). Briefly, gDNA was subjected to enzymatic fragmentation, then the sample library was amplified and purified, and the multiplexed DNA sample library pool was hybridized to enrichment probes. We designed a custom endometrial tumor amplicon panel to cover 22 genes with the highest mutation frequencies specific for EC (list of genes: POLE, POLD1, PTEN, PIK3CA, TP53, CTNNB1, KRAS, NRAS, HRAS, AKT1, EGFR, FGFR2, FBXW7, RB1, ATM, APC, ARID1A, ARID5B, PIK3R1, CDKN2A, PPP2R1A, RPL22) [9]. The genomic target regions were designed to cover all coding exons and all known hotspot loci localized outside the exons. For target enrichment, we designed hybridization probes using HyperDesign tool (Roche), which is an intuitive, user-friendly interface that combines KAPA Target Enrichment technology with KAPA HyperCap Probe and KAPA HyperPETE Primer designs to achieve the best possible coverage of regions of interest, with an estimated coverage of 98.1 %. Sequencing reads from sequencing of custom endometrial tumor amplicon panel were evaluated for quality control by FastQC [10] and aligned to reference genome hg38.p14 [11] by TopHat2 [12]. BAM files which were obtained were sorted (by samtools [13]) and used for detecting of variants in DNA by VarScan2 [14]. Detected variants were used for statistical analysis between defined groups of samples.
Statistical analyses
Standard descriptive statistics - absolute and relative frequencies for categorical variables and median and interquartile range (IQR) for continuous variables - were used to summarize the data. Comparisons of frequencies or distributions between the EC, EIN and control groups were made using Pearsonās Chi-squared test and Kruskal-Wallis rank sum test, respectively. To assess the association between the presence of specific mutations and disease status, univariate logistic regression analysis was performed. Any p-values presented are considered nominal in nature and no adjustment for multiplicity has been done. The analysis was performed using the R software (version 4.3.2). All tests were set as two-sided and tested at 5% significance level.
Data obtained from NGS were processed using the maftools (v2.18.0) package. Only mutations occurring in exons that resulted in protein-level changes were included in the analysis; thus, alterations in ncRNA, splicing regions, 3“UTR, 5“UTR, and synonymous mutations were excluded. Benign and likely benign mutations defined according to NCBI ClinVar database [15] were also excluded. Thus, clinically significant mutations and variants of uncertain significance (VUS) were included in the analysis. Finally, duplicate mutations were excluded during data processing. The occurrence of mutations is presented at the gene level.
Results
Patientsā characteristics
We sequenced and analysed 257 lavage samples from patients enrolled in the study. Of these, 89 samples were histologically confirmed to have endometrial cancer (EC), 80 were from patients with endometrial intraepithelial neoplasia (EIN), and 88 were from the control group. Among these samples, 100 were free of any significant pathogenic mutations across the 22 sequenced genes, while exon mutations in one or more of these genes were detected in 157 samples (TableĀ 1). Overall, we observed a similar frequency of all mutations in the 22 selected genes between the control and EIN groups, while a lower frequency was observed in the EC group. TableĀ 1 also presents the associations between clinicopathological parameters and EC malignancy status. Women in the control group were significantly younger and had a lower BMI compared to those in the EIN and EC groups (pā<ā0.001 and pā=ā0.006, respectively). This finding suggests that age alone is unlikely to be the primary factor driving mutation accumulation, as one would expect an older population to exhibit a higher mutation frequency. Significant differences were also observed in the use of hormonal contraceptives (pā=ā0.001), prevalence of hypertension (pā=ā0.002), and presence of peroral antidiabetic drugs (PAD) (pā=ā0.007).
Analysis of mutations in DNA isolated from lavage fluid collections
Of the 257 uterine lavage samples that were analysed, at least one mutation was detected in 157 cases, and a total of 641 mutations were identified at 466 unique sites in 22 genes that were analysed. The mutation frequencies in each group are presented in Fig.Ā 1.
Comparison of mutation prevalence between different groups of patients
We performed a pairwise analysis of controls vs. EIN, controls vs. EC, and EIN vs. EC. However, the frequency of any of the identified mutations was not significant enough within the control and EIN groups to distinguish the presence of EIN from controls (Fig.Ā 2a). In fact, when comparing the mutation incidence between controls and ECs (Fig.Ā 2b), the opposite trend was observed, with mutations in the KRAS and TP53 genes occurring significantly more frequently in the control group than in patients diagnosed with ECs. Similarly, when comparing EIN vs. EC, mutations in the KRAS gene were observed significantly more frequently in EIN (Fig.Ā 2c). Notably, the mutation frequency of the KRAS gene was significantly higher in the control and EIN groups compared to the EC group (pā=ā0.007 and pā=ā0.002, respectively), further supporting this trend.
Discussion
A key role in cancer elimination is given to cancer prevention. However, most cases of EC are diagnosed in symptomatic women, and there is currently no reliable screening tool to identify high-risk individuals suspected of having EC [16]. However, the group of women who are overweight, have hereditary non-polyposis colon cancer, Lynch syndrome, or have had tamoxifen treatment would benefit from an effective screening strategy. Annually performed clinical examination and transvaginal ultrasound are insufficient. On the other hand, an additional endometrial biopsy and outpatient hysteroscopy could improve screening results, but are not well tolerated and acceptable by all women. Pipelle sampling can be used only in cases with a non-representative biopsy specimen or cervical stenosis [5]. Therefore, we focused on targeted sampling using uterine fluid lavage considered a minimally invasive sampling procedure that, in conjunction with molecular testing, could be useful in both diagnosis and screening. This straightforward and cost-effective method is well-tolerated by women and could be more widely used not only in symptomatic patients, but also in asymptomatic women with an adverse medical history.
An ideal screening method for identifying high-risk patients with EC should be accurate, cost-effective, patient-friendly and at the same time to reliably identify cases requiring invasive testing while providing reassurance to low-risk women. Minimally invasive biofluid sampling has improved early gynecologic cancer detection by enabling the identification of cancer-specific genomic biomarkers, especially in blood, uterine lavage, and cervicovaginal fluid [17]. Examinations of uterine fluid lavages concerning the presence of EC are well-established, as demonstrated by various studies. For instance, a comprehensive genomic analysis of uterine lavage fluid has been shown to detect early endometrial cancers and reveal prevalent driver mutations even in women without histopathologic evidence of cancer [18, 19]. Moreover, recent advancements in NGS have enabled a more precise identification of oncogenic mutations in uterine lavage fluid, highlighting its potential as a liquid biopsy tool for early detection. Targeted molecular analysis of uterine lavage fluid has identified oncogenic mutations that precede clinical symptoms, underscoring the potential for early cancer screening [20]. For example, Chao et al. demonstrated that massively parallel sequencing of uterine lavage specimens successfully detects tumor-associated mutations, allowing risk stratification of patients before conventional histopathological confirmation [21]. In addition, a study by Weng et al. explored the role of circulating free DNA (cfDNA) in uterine lavage fluid, revealing that cfDNA mutations correlate with early-stage EC progression, further strengthening the case for non-invasive molecular diagnostics [22]. Accordingly, Mayo-de-Las-Casas et al. showed that the detection of somatic mutations in peritoneal lavages and plasma of EC patients can be used as a diagnostic tool, offering a broader perspective on the molecular landscape of endometrial cancer [23].
As a proof-of-concept study, we prospectively collected a cohort of 257 women to perform a genetic analysis of DNA extracted from uterine fluid lavages, aiming to identify specific mutations or mutation patterns indicative of endometrial cancer or precancer. However, statistical analysis revealed no significant association between the presence of these mutations and the occurrence of cancer or precancer. These findings highlight inherent limitations and contextual challenges associated with this diagnostic approach. For instance, Nair et al. reported that while uterine lavage fluid could detect cancer-associated mutations in patients with endometrial cancer, similar mutations were present in nearly half of individuals without histopathologic evidence of cancer, raising concerns about specificity and clinical applicability [19]. Similarly, Maritschnegg et al. demonstrated the detection of mutations in uterine lavage fluid but noted inconsistencies in identifying some early-stage cancers, particularly certain subtypes [18]. Moreover, mutations associated with cancer have also been found in benign conditions, further complicating diagnostic accuracy. Genetic heterogeneity within tumors also poses a challenge; Mota et al. highlighted that intra-tumor heterogeneity may lead to under-detection of mutations when relying solely on uterine fluid samples [24]. These issues underscore the need for more refined approaches to enhance the specificity and reliability of uterine lavage as a diagnostic tool.
In conclusion, although genetic analysis of uterine fluid lavage could be a promising tool for early detection of EC, we did not confirm this trend in our study. This may be due to the limited specificity of this approach, or its sensitivity or potential overlap with benign conditions, which ultimately highlights the need for complementary diagnostic tools and more thorough validation. Other emerging factors warrant further investigation for their potential in the early detection of EC. These include copy number variation (CNV) analysis, which provides valuable insights into genomic alterations associated with EC [25]; and the identification of specific gene methylation patterns, which have demonstrated diagnostic accuracy comparable to endometrial biopsy and have been validated in prospective studies [26]. In addition, the role of the gut microbiome is an intriguing and emerging field of research. Some studies have suggested that specific bacterial species, such as Porphyromonas somerae, may have predictive value, particularly in postmenopausal and obese patients [27, 28]. However, no study has yet conclusively demonstrated that the microbiome alone has the same diagnostic potential as other molecular markers, and further validation is needed before it can be considered a reliable clinical tool. Integrating these biomarkers into a multifaceted diagnostic framework could enhance early detection strategies, improve risk assessment, and ultimately lead to better patient outcomes.
Limitations
While targeted sampling using uterine fluid lavage combined with genetic analysis of selected genes has been investigated as a potential approach for detecting endometrial cancer, our findings did not identify a specific mutational pattern, and even mutational load alone did not differentiate cases from controls. This highlights important limitations, including the possibility that genetic alterations detected in the lavage may not always indicate malignancy, leading to false positives, while tumors lacking mutations in the selected genes can result in false negatives. Additionally, the genetic diversity of endometrial cancer and population-specific variations in baseline genetic markers further complicate interpretation, as a limited gene panel may fail to capture the full spectrum of relevant mutations, potentially missing key diagnostic markers.
Data availability
The experimental data that support the findings of this study are available in NCBI database as BioProject PRJNA1196106 and can be also accessible via link: http://www.ncbi.nlm.nih.gov/bioproject/1196106.
References
Lortet-Tieulent J, Ferlay J, Bray F, Jemal A. International patterns and trends in endometrial Cancer incidence, 1978ā2013. J Natl Cancer Inst. 2018;110(4):354ā61.
Mazidimoradi A, Momenimovahed Z, Khalajinia Z, Allahqoli L, Salehiniya H, Alkatout I. The global incidence, mortality, and burden of uterine cancer in 2019 and correlation with SDI, tobacco, dietary risks, and metabolic risk factors: an ecological study. Health Sci Rep. 2024;7(1):e1835.
Sangtani A, Wang C, Weaver A, Hoppman NL, Kerr SE, Abyzov A, et al. Combining copy number, methylation markers, and mutations as a panel for endometrial cancer detection via intravaginal Tampon collection. Gynecol Oncol. 2020;156(2):387ā92.
Hrstka R, Podhorec J, Nenutil R, Sommerova L, Obacz J, Durech M, et al. Tamoxifen-Dependent induction of AGR2 is associated with increased aggressiveness of endometrial Cancer cells. Cancer Invest. 2017;35(5):313ā24.
Morice P, Leary A, Creutzberg C, Abu-Rustum N, Darai E. Endometrial cancer. Lancet. 2016;387(10023):1094ā108.
Spira A, Yurgelun MB, Alexandrov L, Rao A, Bejar R, Polyak K, et al. Precancer atlas to drive precision prevention trials. Cancer Res. 2017;77(7):1510ā41.
Schlosshauer PW, Ellenson LH, Soslow RA. Beta-catenin and E-cadherin expression patterns in high-grade endometrial carcinoma are associated with histological subtype. Mod Pathol. 2002;15(10):1032ā7.
Holcakova J, Bartosik M, Anton M, Minar L, Hausnerova J, Bednarikova M et al. New trends in the detection of gynecological precancerous lesions and Early-Stage cancers. Cancers (Basel). 2021;13(24).
Cancer Genome Atlas Research N, Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, et al. Integrated genomic characterization of endometrial carcinoma. Nature. 2013;497(7447):67ā73.
Andrews S. FastQC: A Quality Control Tool for High Throughput Sequence Data. http://www.bioinformaticsbabrahamacuk/projects/fastqc/. 2010.
Cunningham F, Allen JE, Allen J, Alvarez-Jarreta J, Amode MR, Armean IM, et al. Ensembl 2022. Nucleic Acids Res. 2022;50(D1):D988ā95.
Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14(4):R36.
Danecek P, Bonfield JK, Liddle J, Marshall J, Ohan V, Pollard MO et al. Twelve years of samtools and BCFtools. Gigascience. 2021;10(2).
Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22(3):568ā76.
Landrum MJ, Lee JM, Riley GR, Jang W, Rubinstein WS, Church DM, et al. ClinVar: public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res. 2014;42(Database issue):D980ā5.
Asaturova A, Zaretsky A, Rogozhina A, Tregubova A, Badlaeva A. Advancements in minimally invasive techniques and biomarkers for the early detection of endometrial cancer: A comprehensive review of novel diagnostic approaches and clinical implications. J Clin Med. 2024;13:24.
Shen Y, Yang W, Liu J, Zhang Y. Minimally invasive approaches for the early detection of endometrial cancer. Mol Cancer. 2023;22(1):53.
Maritschnegg E, Wang Y, Pecha N, Horvat R, Van Nieuwenhuysen E, Vergote I, et al. Lavage of the uterine cavity for molecular detection of mullerian duct carcinomas: A Proof-of-Concept study. J Clin Oncol. 2015;33(36):4293ā300.
Nair N, Camacho-Vanegas O, Rykunov D, Dashkoff M, Camacho SC, Schumacher CA, et al. Genomic analysis of uterine lavage fluid detects early endometrial cancers and reveals a prevalent landscape of driver mutations in women without histopathologic evidence of cancer: A prospective Cross-Sectional study. PLoS Med. 2016;13(12):e1002206.
Martignetti JA, Pandya D, Nagarsheth N, Chen Y, Camacho O, Tomita S et al. Detection of endometrial precancer by a targeted gynecologic cancer liquid biopsy. Cold Spring Harb Mol Case Stud. 2018;4(6).
Chao A, Wu KY, Lin CY, Lee YS, Huang HJ, Tang YH, et al. Towards less invasive molecular diagnostics for endometrial cancer: massively parallel sequencing of endometrial lavage specimens in women attending for an office hysteroscopy. J Mol Med (Berl). 2022;100(9):1331ā9.
Weng CH, Wu KY, Wang CJ, Huang HJ, Tsai CL, Lin CY, et al. Massively parallel sequencing of endometrial lavage specimens for the detection of cancer-associated mutations in atypical and non-atypical endometrial hyperplasia. Front Med (Lausanne). 2022;9:1090788.
Mayo-de-Las-Casas C, Velasco A, Sanchez D, Martinez-Bueno A, Garzon-Ibanez M, Gatius S, et al. Detection of somatic mutations in peritoneal lavages and plasma of endometrial cancer patients: A proof-of-concept study. Int J Cancer. 2020;147(1):277ā84.
Mota A, Colas E, Garcia-Sanz P, Campoy I, Rojo-Sebastian A, Gatius S, et al. Genetic analysis of uterine aspirates improves the diagnostic value and captures the intra-tumor heterogeneity of endometrial cancers. Mod Pathol. 2017;30(1):134ā45.
Dugo E, Piva F, Giulietti M, Giannella L, Ciavattini A. Copy number variations in endometrial cancer: from biological significance to clinical utility. Int J Gynecol Cancer. 2024;34(7):1089ā97.
Zheng H, Yu C, Yang L, Zhou F, Liu A. Research progress of DNA methylation markers for endometrial carcinoma diagnosis. J Cancer. 2025;16(3):812ā20.
Di Tucci C, De Vito I, Muzii L. Immune-Onco-Microbiome: A new revolution for gynecological cancers. Biomedicines. 2023;11(3).
Zheng W, Lin X, Chen H, Yang Z, Zhao H, Li S, et al. Gut microbiota and endometrial cancer: research progress on the pathogenesis and application. Ann Med. 2025;57(1):2451766.
Acknowledgements
Special thanks to RECAMO staff for their help in the preanalytical processing of uterine lavages.
Funding
The work was supported by CHRC - project NU21-09-00031, by the project BBMRI.cz no. LM2023033, MH CZ - DRO (MMCI, 00209805) and by the project SALVAGE (P JAC; reg. no. CZ.02.01.01/00/22_008/0004644)ā funded by the European Union and by the State Budget of the Czech Republic.
Author information
Authors and Affiliations
Contributions
R.H. designed experiments and wrote the main manuscript text; F.Z.K. was responsible for bioinformatics analysis of sequencing data; L.M., M.A., D.M., M.B., P.B. and L.M. conducted the clinical study and made several contributions to the paper; T.K. and M.S. performed the laboratory analysis; P.O. executed the statistical analysis; J.H. was responsible for histological verifications and participated on the manuscript writing; V.W. designed experiments, secured funding and participated on the manuscript writing. Each author has reviewed and consented to their contribution as detailed in the submission.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The study was conducted in accordance with the Declaration of Helsinki and received ethical approval from the Ethics Committee of the University Hospital in Brno, Czech Republic (protocol number 03-100620/EK) for research involving human participants. All participants provided written informed consent prior to their participation in the study.
Consent for publication
Not Applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisherās note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the articleās Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleās Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Hrstka, R., Zavadil-Kokas, F., Moukova, L. et al. Genetic analysis of uterine lavage fluids to identify women at high risk of endometrial cancer. BMC Res Notes 18, 117 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07173-8
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07173-8