Open Access

Potential role of Fanconi anemia pathway in the pathogenesis of endometrial cancer (Review)

  • Authors:
    • Mengmeng Yao
    • Chuqi Liu
    • Huiyu Ping
    • Kaidi Meng
    • Xinru Li
    • Qingxin Li
    • Yuanmin Qi
    • Ziming Zhu
    • Li Zhang
    • Aizhong Han
  • View Affiliations

  • Published online on: August 21, 2025     https://doi.org/10.3892/mmr.2025.13660
  • Article Number: 295
  • Copyright: © Yao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endometrial cancer (EC) is a common gynecologic malignancy that often exhibits molecular features such as extensive somatic copy number alterations, microsatellite instability and frequent TP53 mutations, which considerably affect the physical and mental well‑being of women. The Fanconi anemia (FA) pathway is a DNA damage repair pathway involving multiple FA genes that play crucial roles in DNA damage repair as well as the maintenance of genome stability. Abnormalities in FA, such as deletions or mutations, may lead to defects in DNA damage repair, resulting in increased genomic instability and/or an abnormal cell cycle, ultimately leading to EC. This comprehensive review provides a systematic summary of EC‑related FA genes, elucidates the roles of various FA genes in EC and further speculates on their related mechanisms to facilitate the development of targeted therapies that specifically target key genes, leading to a more accurate and efficient treatment for EC. The present review searched PubMed and Google Scholar for articles published in English up to June 2025 using keywords such as Fanconi anemia pathway, 22 FA genes (FANCA, FANCB, FANCC, FANCD1/BRCA2, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCJ/BRIP1, FANCL, FANCM, FANCN/PALB2, FANCO/RAD51C, FANCP/SLX4, FANCQ/XPF, FANCR/RAD51, FANCS/BRCA1, FANCT/UBE2T, FANCU/XRCC2, FANCV/REV7, FANCW/RFWD3), endometrial cancer (type I: Endometrioid adenocarcinoma; Type II Uterine serous carcinoma, clear‑cell carcinoma, carcinosarcoma), somatic copy number alterations, microsatellite instability, TP53 mutations, pathogenesis, genomic instability, target therapy.

Introduction

Endometrial cancer (EC) is one of the most prevalent gynecological malignancies worldwide (1); its global incidence has increased annually over the past 20 years. Although >50% of women diagnosed with EC are aged 50–69 years, the diagnosis rate in women aged <50 years has shown an increasing trend. Although the overall mortality rate has slightly decreased in recent years, EC remains one of the leading causes of cancer-related deaths among postmenopausal women in a number of countries (24). Traditionally, EC has been classified into two main types: Endometrioid (type I) and non-endometrioid (type II). Type I is a low-grade tumor associated with high estrogen levels, with the histological type of endometrioid adenocarcinoma (EAC) accounting for ~80% of all EC cases. Of the patients, 95% have a good prognosis. By contrast, type II is a highly aggressive, high-grade tumor, including serous carcinoma of the uterus, clear cell carcinoma and carcinosarcoma, with a poor prognosis (59). Type II carcinomas and 25% of high-grade type I carcinomas exhibit extensive somatic copy number alterations (SCNA), frequent TP53 mutations and other molecular manifestations, whereas most type I carcinomas exhibit microsatellite instability (MSI) (10), indicating that genomic instability and cell cycle abnormalities may be closely related to the development of EC.

Normal function and growth of cells depend on the orderly progression of the cell cycle. In both unicellular and multicellular eukaryotes, the cell cycle is regulated by a complex network of regulatory mechanisms (11). In this complex network, genes involved in the DNA damage repair pathway play an important role and abnormalities in these genes may affect endometrial cell genome stability and/or lead to the failure of cell cycle regulation, increased susceptibility to EC and even ultimately trigger EC. The Fanconi anemia (FA) pathway plays a crucial role in DNA interstrand cross-linking (ICL) repair and the maintenance of genomic integrity (12). FA genes participate in DNA damage response (DDR) through the FA and homologous recombination (HR) pathways (13). FA gene deletions or variants may lead to DNA damage repair defects, causing genomic instability such as SCNA and MSI, or cell cycle abnormalities, ultimately leading to the development of EC. The present comprehensive review aimed to summarize the association between FA genes and EC occurrence, explore the potential relationship, open new avenues for future research on EC and FA genes and provide potential insights into the treatment of EC.

Mechanisms of FA

FA is rare a chromosomal disorder characterized by genomic instability and an increased susceptibility to cancer caused by mutations in the FA gene. By contrast, the FA pathway, a DNA damage repair pathway involving multiple FA genes, is crucial for normal DNA repair processes and maintenance of genomic stability (14). To date, 22 genes involved in the FA pathway have been identified and can be categorized into upstream, midstream and downstream components. The upstream genes include the lesion recognition gene FANCM, FA core complex-related genes (FANCA, FANCB, FANCC, FANCE, FANCF, FANCG and FANCL) and the E2 ubiquitin ligase-related gene FANCT/UBE2T. Midstream genes include FANCD2 and FANCI, which form the FANCI-FANCD2 (ID2) complex. The downstream genes included HR repair-related genes (FANCD1/BRCA2, FANCJ/BRIP1, FANCN/PALB2, FANCO/RAD51C, FANCR/RAD51, FANCS/BRCA1, FANCU/XRCC2 and FANCW/RFWD3), nucleolytic incision-related genes (FANCQ/XPF and FANCP/SLX4) and trans-lesion synthesis (TLS)-related genes FANCV/REV7 (15). The entire FA core complex consists of seven FA proteins (FANCA, FANCB, FANCC, FANCE, FANCF, FANCG and FANCL) and FANCA-associated peptides FAAP100, FAAP24 and FAAP20 (16,17). FAAP24 can target FANCM to form the FANCM-FAAP24 complex, recognize ICL and promote the recruitment of the FA core complex to DNA damage sites to participate in DNA damage repair (18). This core complex has E3-ubiquitin ligase activity and is essential for DNA damage repair by mediating mono-ubiquitination of the ID2 complex in response to DNA damage (15). The ID2 complex plays a critical role in the FA pathway and is recruited to the ICL site before FANCD2 mono-ubiquitination, thus participating in pathway activation (19). Additionally, downstream FA genes coordinate the three key DNA repair processes: Nucleolytic incision, TLS and HR, all of which are essential for ICL repair (20). The FA pathway plays a key role in DNA damage repair and participates in DNA damage repair and the maintenance of genome stability by interacting with other DNA repair pathways. Defective DNA repair increases genomic instability, which may lead to cancer development. Several FA genes are closely associated with EC (21). To elucidate the roles of FA genes in EC, the upstream (Table I), midstream (Table II) and downstream genes (Table III) of the EC-associated FA pathway were considered entry points and the underlying mechanisms contributing to EC development and progression were explored.

Table I.

Functional evidence of upstream FA gene in EC.

Table I.

Functional evidence of upstream FA gene in EC.

GeneFunctionExperimental evidencesPotential mechanismEC subtypes
FANCEFA core complexGenetic testing (24)MSI (26)Type I EC (EAC)
Cell model (25)Cell cycle regulation (25,27)
FANCCFA core complexGenetic testing (29)DNA replication fork stagnation (30)EC
Mouse model (33)Cell cycle regulation (31,33)
FANCAFA core complexGenetic testing (3841)MSI (38)Type I EC
Clinical patients (3841)MMR (37)

[i] FA, Fanconi anemia; EC, endometrial cancer; MSI, microsatellite instability; MMR, mismatch repair; EAC, endometrioid adenocarcinoma.

Table II.

Functional evidence of midstream FA gene in EC.

Table II.

Functional evidence of midstream FA gene in EC.

GeneFunctionExperimental evidencesPotential mechanismEC subtypesClinical significance
FANCD2ID2 complexCell model (43)MSI (44)Type I ECEvaluating prognosis
Clinical patients (9)Cell cycleType II EC
regulation (43)
FANCIID2 complexCancer GenomeDownregulation ofType II ECEvaluating prognosis
Atlas (49, 50)TP53 expression (47)(UC)
Clinical patients (4951)SCNA (49,50)
HR repair defects (51)

[i] FA, Fanconi anemia; EC, endometrial cancer; MSI, microsatellite instability; SCNA, somatic copy number alterations; ID2, FANCI-FANCD2; HR, homologous recombination; UC, uterine cancer.

Table III.

Functional evidence of downstream FA gene in EC.

Table III.

Functional evidence of downstream FA gene in EC.

GeneFunctionExperimental evidencePotential mechanismEC subtypes
FANCNHR repairGenetic testing (62)SCNA (66)Type I EC (EAC)
Clinical patients (6365)TP53/Cell cycle regulation (67)
Mouse model (67)
FANCOHR repairGenetic testing (64,71)HR repair defects (63)Type II EC (UC)
Cell cycle regulation (68)
FANCJHR repairGenetic testing (60,76,78)HR repair defects (72)Type I EC
Clinical patients (71,77)Cell cycle regulation (75)Type II EC
Mouse model (79)MSI (79)
FANCS/HR repairCohort study (84)HR repair defects (77)Type I EC
FANCD1 Genetic testing (77,85,86)TP53/Cell cycleType II EC (UC)
Clinical patients (77,85,86)regulation (82,83)
MSI (86)
SCNA (86)
FANCRHR repairClinical patients (88)HR repair defects (88)Type II EC
FANCUHR repairGenetic testing (98,99)HR repair defects (96,97)Type I EC (EAC)
Clinical patients (98,99)MSI (99)
FANCWHR repairGenetic testing (21)HR repair defects (102)EC
Cell cycle regulation (103,104)

[i] FA, Fanconi anemia; EC, endometrial cancer; HR, homologous recombination; SCNA, somatic copy number alterations; MSI, microsatellite instability; EAC, endometrioid adenocarcinoma; UC, uterine cancer.

FA genes and EC occurrence

Upstream FA genes and EC
i) FANCE

FANCE encodes a 58 kDa nuclear protein and its products are components of the FA core complex. The integrity of this complex is essential for DNA damage repair and genomic stability (22,23). Using CBioportal data analysis, Salomão et al (21) found that the FANCE mutation rate in EC was 4.54%. Lynch syndrome (LS) is a common hereditary cancer that is associated with EC. de Angelis de Carvalho et al (24) incidentally identified a variant of the FANCE gene (c.929dupC; p.Val311Serfs*2-LP) during germline polygenic testing in 14 patients. Zheng et al (25) found that FANCE expression was markedly lower in EAC than in normal endometrium and that reduced expression was associated with older age and higher tumor grade. By constructing FANCE-overexpressing EC cells, we found that FANCE overexpression promoted the repair of ICL and double-strand breaks (DSBs) in EC. However, in contrast to the findings of Zheng et al (25), Zhou et al (26) analyzed the FANCE gene in 33 types of cancer, including EC and found that high FANCE expression was markedly positively associated with MSI, particularly in EC. This contradiction may be due to interference from different subtypes of EC (EAC compared with mixed subtypes) and the possible biphasic function of FANCE in EC. In the early stages, it repairs DNA damage and acts as a tumor suppressor, whereas in the late stages, its continued high expression causes erroneous repair, leading to MSI and acting as a tumor promoter. Zheng et al (25) found that FANCE is involved in cell cycle regulation. Overexpression of FANCE leads to S phase delay and G2/M phase arrest in EC cells, thereby inhibiting the growth of EC tumors in nude mice, whereas low expression induces genomic instability and promotes EC development. Furthermore, Lin et al (27) found that FANCE may participate in regulating squamous cell carcinoma cell cycle through the Wnt/β-catenin signaling pathway, but whether this mechanism applies to EC needs to be further verified in EC models. In summary, abnormal expression or mutation of FANCE may lead to EC by affecting DNA damage repair, cell cycle, microsatellite stability and other factors. However, there are still contradictions regarding the direction of action and the molecular mechanisms of this gene in EC. Future studies are needed to distinguish EC subtypes further and to verify their functions and specific molecular mechanisms in different EC subtypes.

ii) FANCC

FANCC is a key component of the FA core complex that promotes two DNA damage repair processes, HR repair and TLS, which are essential for maintaining genomic stability (28). Research has found that the mutation rate of the FANCC gene in EC is 5.1 (21). EC is one of the most common uterine cancers (UC). In a study by Heald et al (29), four pathogenic or possibly pathogenic germline variants of the FANCC gene were identified by genetic testing of 953 patients with UC, indicating that variants in this gene may increase the risk of EC. FANCC gene deletion leads to stalling of the DNA replication fork, thereby increasing genomic instability (30), which drives EC development. Additionally, FANCC is involved in cell cycle regulation (31). FANCC mutations result in DNA damage repair defects, increased spontaneous chromosome breaks and G2/M phase arrest (28,32). Li et al (33) found that FANCC-deficient mice exhibited cell cycle abnormalities that promoted the malignant progression of endometrial cells. The FANCC gene is associated with multiple FA genes. For example, FANCC and FANCG jointly participate in the monoubiquitination of FANCD2 (32). Additionally, FANCC forms a complex with FANCE and FANCF (the FANCC-FANCE-FANCF complex) to regulate meiotic recombination (34). However, this functional synergistic mechanism has not been explored in EC models. Whether the FANCC gene has a specific effect on different EC subtypes requires further investigation.

iii) FANCA

As a key component of the FA core complex (13), FANCA plays a crucial role in the repair of DSBs and ICL through the HR repair pathway in the S/G2 phase of cells, which is essential for maintaining genome stability (35,36). In addition, FANCA participates in the mismatch repair (MMR) process by enhancing the interactions between MSH2 and MLH1 (37). Salomão et al (21) discovered a high mutation rate of FANCA in the upstream FA gene in EC, with mutation rates as high as 8.88%. Drusbosky et al (38) analyzed genomic data from 1,988 patients, of whom 12% exhibited high levels of MSI and a small number of patients were found to have pathogenic FANCA germline mutations. Kral et al (39) conducted germline genetic testing on 527 patients, of whom 284 had type I cancer and found that FANCA was a candidate susceptibility gene for EC. In addition, Drusbosky et al (38) and Liu et al (40) found that somatic mutations in the FANCA gene are also associated with EC. Atypical endometriosis (A-EMS) is associated with endometrial intraepithelial neoplasia. Recently, Wepy et al (41) revealed that 25% of 36 patients with A-EMS had concurrent EMS-related tumors, such as ipsilateral endometrioma, serous tumors, or clear cell tumors. A frameshift mutation in the FANCA gene (p.I865Kfs*20) was detected by second-generation sequencing in six patients with A-EMS. Although multiple studies (21,3841) have suggested an association between FANCA abnormalities and EC, the specific mechanisms by which FANCA mutations contribute to EC remain unclear. According to current research, mutations in this gene may impair DNA damage repair by affecting the FA pathway or MMR, thereby increasing the risk of EC. Future studies should further refine EC subtypes to investigate the role of this gene and its specific mechanisms of action across different EC subtypes.

Midstream FA genes and EC
ID2 ubiquitination-related genes
i) FANCD2

FANCD2 plays a key role in DNA damage repair by interacting with FANCI to form the ID2 complex, which is critical for activating the FA pathway (42). Research has shown that the mutation rate of FANCD2 in EC is 9.07% (21). In addition, immunohistochemical results show that compared with normal tissue and atypical hyperplasia tissue, the expression level of FANCD2 is markedly upregulated in type I EC tissue. Knockdown of FANCD2 can lead to cell cycle arrest in the G1 phase in EC cell lines, thereby inhibiting their proliferation and migration of EC cells (43). At the same time, Mhawech-Fauceglia et al (9) evaluated the expression of various DNA repair proteins in tumor cells from 357 EC patients (of these, 262 cases were type I EC, 76 cases were type II EC and 19 cases were carcinosarcoma) and found that FANCD2 overexpression was markedly associated with lymphatic invasion and the recurrence of high-grade EC. Cox regression and Kaplan-Meier analyses showed that high expression of FANCD2 was associated with poor overall survival in patients with EC. In addition, a study also found that FANCD2 expression positively associated with MSI in EC (44). This suggests that the overexpression of FANCD2 may lead to the accumulation of MSI in endometrial cells, leading to genomic instability and eventually to the occurrence of EC. After FANCD2 knockdown, the sensitivity of EC cells to paclitaxel, cisplatin and doxorubicin increased markedly, suggesting that FANCD2 promotes EC cells (45), highlighting its prognostic value in EC. In summary, high expression of FANCD2 may increase the risk of EC by affecting DNA damage repair and cell genome stability. In addition, overexpression of this gene is closely related to the poor prognosis of EC and FANCD2 may be a valuable biomarker for evaluating the prognosis of patients with EC.

ii) FANCI

FANCI forms a heterodimer with FANCD2, which is then recruited to the site damaged by related proteins, thereby playing an important role in DNA damage repair (46). The absence of FANCI markedly downregulates the expression of core FA proteins such as FANCD2 and FANCB, leading to the accumulation of DNA damage within cells. Additionally, FANCI silencing partially reduced TP53 expression, causing cell cycle arrest at the G1 phase (47). Research has shown that the mutation rate of FANCI in EC is 8.70% (21). This is consistent with the observation by Zhao et al (48) that the mutation rate of the FANCI gene in EC exceeded 8%. Pan-cancer research using The Cancer Genome Atlas (TCGA) has found that somatic mutations in FANCI exist in multiple types of cancer. Of the 517 EC, 43 cases (8.32%) harbored mutations. In addition, Fierheller et al (49) reported a case of ovarian cancer carrying FANCI c.1813C > T in a patient with extensive genome-wide SCNA (50). Dong et al (51) identified FANCI mutations in the HR repair gene mutation spectrum of patients with uterine serous carcinoma. This suggests that type II EC are associated with HR repair defects caused by FANCI mutations. In addition, Bi et al (52) analyzed the TCGA dataset of patients with EC and found that FANCI overexpression may be associated with a poor prognosis in EC. FANCI may become a valuable biomarker for assessing the prognosis of patients with EC. Further research is needed to investigate the effect of FANCI on type I EC and the regulatory role of TP53 in developing EC to clarify the function and underlying mechanisms of this gene in different EC subtypes and guide treatment strategies.

Downstream FA genes and EC
HR repair-related genes and EC
i) FANCN

FANCN, also known as PALB2, is closely associated with BRCA2 (53). The coiled-coil, WD40 and ChAM domains of PALB2 ensure the normal formation of RAD51 filaments and promote the correct binding of RAD51 to DNA, which is critical for the HR repair of DSBs (54). In addition, PALB2 interacts with BRCA1/2 to form the BRCA1-PALB2-BRCA2 axis, which promotes DSB repair, maintains genomic stability and is crucial for inhibiting tumor progression (55). Multiple studies have identified mutations that cause PALB2 loss of function in patients (5661). Salomão et al (21) analyzed CBioportal data and found that the mutation rate of FANCN in EC was 6.62%. Johnatty et al (62) determined the role of PALB2 mutations in increasing the risk of EC using a multigene panel detection. In 2019, one patient with EC with a pathogenic PALB2 mutation was identified among 198 women in China (63). Sequencing analysis based on the TCGA-UCEC cohort revealed that the mutation frequency of the PALB2 gene in EC cases was markedly higher than that in the general population, more than doubling (0.32 vs. 0.14%). Researchers also identified a patient with a pathogenic PALB2 frameshift mutation (c.3116delA, p.Asn1039Ilefs) who was diagnosed with stage I endometrioid EC at 70 years of age (64). A PALB2 gene mutation [NM_024675.3 (PALB2): c. (3113+1_3114-1)_ (3201+1_3202-1) del] was identified in the tumor tissue of a patient with serous EC; this mutation is considered pathogenic (65). In addition, Foo et al (66) used whole-exome sequencing (WES) to discover that PALB2 mutations in breast cancer patients can lead to multiple genomic SCNA. This suggests that the gene mutation may be associated with type II EC; however, further validation in patients with EC is required. Bowman-Colin et al (67) found that PALB2 coordinates with the cell cycle factor Trp53 to inhibit breast cancer formation in mice, suggesting that PALB2 mutations affect TP53 function and lead to cell cycle abnormalities. In summary, PALB2 mutations are associated with both type I and type II EC; however, further investigation is needed into the specific pathways through which this gene regulates HR repair in endometrial cells and its impact on endometrial cell cycle regulation.

ii) FANCO

FANCO, also known as RAD51C, is a tumor suppressor gene. As a paralog of RAD51, this gene plays a central role in the HR repair of ICLs and DSBs by forming RAD51B-RAD51C-RAD51D-XRCC2 and RAD51C-XRCC3 complexes, which are critical for maintaining replication fork integrity (6870). Additionally, this gene participates in the regulation of DNA damage-induced cell cycle S-phase checkpoint by activating the phosphorylation of CHK2 (68). Salomão et al (21) analyzed CBioportal data and found that the mutation rate of FANCO in EC was 3.21%. Ring et al (71) detected three cases of serous EC carrying RAD51C mutations among 381 EC patients using a multi-gene panel, suggesting that mutations in this gene may be associated with type II EC (serous carcinoma). Kondrashova et al (64) analyzed 30 known and candidate EC risk genes in patients with EC (TCGA-UCEC study) and the general population (gnomAD database) using tumor sequencing data and reported a nine-fold increase in the frequency of pathogenic or possible pathogenic variations in the HR repair-related gene RAD51C (0.97 vs. 0.1%). This study underscores the potential role of RAD51C in EC development and highlights its potential as a candidate gene for the future exploration of EC risks. In conclusion, clinical genomic data suggest that RAD51C mutation is associated with type II EC (especially serous carcinoma), but its role and related mechanisms in type I EC remain unclear. Additionally, the CHK2 phosphorylation regulatory pathway also needs to be further verified in endometrial cell models.

iii) FANCJ

FANCJ, also known as BACH1 and BRCA1 interacting protein 1 (BRIP1), is a key downstream gene of the FA pathway. This gene can bind to the carboxyl-terminus of BRCA1 to promote HR repair (72). Moreover, this gene can be recruited to DNA break sites and, together with CTLP, promotes DNA end resection and repair of ICL and DSBs (73,74). It also participates in cell-cycle regulation through phosphorylation and dephosphorylation states (75). Salomão et al (21) analyzed data from CBioportal and found that the mutation rate of FANCJ in EC was 7.37%. Heeke et al (60) used NGS600 sequencing to detect an FANCJ mutation rate of 0.14% in 1,475 patients with EC. At the same time, studies have shown that BRIP1 has a higher mutation frequency in type II cancer than in type I cancer (76). Ring et al (71) reported the presence of a harmful germline mutation in BRIP1 in 13 non-LS EC patients (p.Lys705Thrfs*10). de Jonge et al (77) reported BRIP1 mutations c.632delC and P.Ro211fs in two EC patients with HR repair deficits. A frameshift mutation (p.Q554Hfs*35) in FANCJ was identified as pathogenic in high-grade EC (78). In addition, it has been found that FANCJ mutant mice show an increase in spontaneous MSI (79). This suggests that this may be related to type I EC. In summary, clinical genomic data suggest that BRIP1 mutations are associated with EC and that the mutation frequency is higher in type II than in type I. However, the functional mechanism in type I EC remains unclear. Further research is needed to investigate the mechanism of FANCJ-mediated MSI in endometrial cells and the specific role of its phosphorylation in cell cycle regulation.

iv) FANCS and FANCD1

FANCS/BRCA1 and FANCD1/BRCA2 play key roles in DNA damage response through HR repair. BRCA1 is mainly localized at DNA damage sites, where it binds to PALB2 to initiate DNA end excision and promotes RAD51 loading. Additionally, BRCA1, in conjunction with BARD1, enhances the activity of the RAD51 recombinase, whereas BRCA2 protects against stagnant DNA replication bifurcations (80,81). In addition, BRCA1/2 interacts with TP53 to regulate cell cycle progression. Functional deficiency can lead to genomic instability, ultimately promoting cancer occurrence and development (82,83). Studies have shown that FANCS/BRCA1 and FANCD1/BRCA2 mutations are markedly associated with EC. Data analysis from CBioportal showed that the mutation rate of FANCS in EC was 9.26% and that of FANCD1 was 15.31% (21). In a large cohort study of BRCA1/2 mutation carriers, de Jonge et al (84) found that compared with the general population and non-BRCA1/2 gene mutation carriers, BRCA1/2 mutation carriers had a two- to three-fold increased risk of developing EC and BRCA1 gene mutation carriers had the highest risk of developing rare serous and p53-abnormal EC subgroups. A recent study identified 35 patients with BRCA1/2 copy number loss and eight patients with BRCA1/2 somatic mutations among 187 high-grade ECs using shallow whole-genome sequencing (85). In addition, in a study by Smith et al (86), MSI or SCNA pathogenic BRCA1/2 mutations were identified in 13 of 769 patients with high MSI or SCNA levels, whereas TP53 mutations were identified in all patients. de Jonge et al (77) identified pathogenic variants of BRCA1 and BRCA2 in eight ECs with HR repair defects: BRCA1, c.4327C>T, p.Arg1443*; BRCA2, c.6373dupA, p. Thr2125fs c.6306_6413del; and BRCA2, p.Ser2103_Val2138del. In summary, the clinical data suggest that BRCA1/2 mutations are associated with both Type I and Type II EC, with a more significant association with high-grade EC (Type II). However, the mechanisms underlying these mutations in high-risk subtypes, such as serous carcinoma, require further clarification. Additionally, the synergistic regulation of the cell cycle by BRCA1/2 and TP53, as well as the mechanisms related to MSI and SCNA caused by BRCA1/2 mutations, require further validation in endometrial cells.

v) FANCR

FANCR/RAD51 is an essential recombinase involved in HR repair that forms nuclear protein filaments by binding to single-stranded DNA, promoting the pairing of RAD51 with DNA sequences and participating in homologous search and chain exchange, which are essential for the maintenance of HR repair and genome stability (87). Bioportal data analysis shows that the mutation rate of FANCR in EC is 3.40% (21). Auguste et al (88) evaluated the degree of DNA damage in 116 patients with high-grade EC using immunohistochemistry. The results showed that complete deletion of RAD51 resulted in severe DNA damage in 28 patients. In addition, several studies have suggested that RAD51 135G/C and 135C/C polymorphisms may be related to the occurrence and development of EC (8991). Michalska et al (92) suggested that the 135C/C polymorphism may be more closely associated with EC. Additionally, Krupa et al (93) found that the RAD51 C/C genotype was closely associated with the incidence of EC in patients with higher levels of basal DNA damage. In summary, existing clinical data suggest that the absence of FANCR/RAD51 is associated with high-grade EC (Type II), however, the role of this gene in Type I EC and the underlying mechanisms remain unclear. Furthermore, the specific molecular pathways through which RAD51 135G/C and 135C/C polymorphisms influence EC susceptibility require further validation.

vi) FANCU

FANCU, also known as XRCC2, plays a vital role in the HR repair of ICLs and DSBs (94,95). XRCC2 is a potential tumor suppressor gene in mammals. Mutations in this gene can lead to defects in DNA damage repair and SCNA, thereby disrupting genomic stability (96,97). Bioportal data analysis showed that the FANCE mutation rate in EC was 3.02% (21). Nero et al (98) performed genome sequencing of 137 patients with recurrent EC and identified XRCC2 mutations in 2.9% of the patients. Taylor et al (99) identified one XRCC2 gene frameshift mutation case in 50 cases of MSI-H endometrioid adenocarcinoma. Additionally, XRCC2 SNP-41657C/T (rs718282) and 4234G/C (rs3218384) increased EC susceptibility (100,101). In summary, the current study indicated that FANCU/XRCC2 mutations and SNP polymorphisms increase the risk of EC. However, the specific pathogenic mechanisms by which XRCC2 mutations drive the development of endometrioid adenocarcinoma and how SNP polymorphisms in this gene contribute to EC require further clarification. Additionally, the mechanism by which XRCC2 deletion leads to SCNA requires further validation in endometrial cells.

vii) FANCW

FANCW, also known as RFWD3, is an E3 ubiquitin ligase that promotes the timely removal of RPA and RAD51 from DNA damage sites and plays a crucial role in DNA HR repair and maintenance of genome stability (102). RFWD3 is also necessary to maintain a normal cell cycle and participate in the DDR by positively regulating p53. Knockdown of the RFWD3 gene may lead to cell-cycle arrest (103,104). Mutations in the FANCW gene have been identified in various cancers, including EC, ovarian cancer and bladder cancer. Compared with other types of cancer (2.23% in ovarian cancer and 3.89% in bladder cancer), the mutation frequency of this gene in EC is relatively high (4.73%) (21). However, the mechanisms by which RFWD3 regulates the participation of p53 in cell cycle regulation and causes genomic instability due to mutations in endometrial cells require further validation.

Summary and prospect

EC is the most prevalent cancer of the female reproductive tract and its incidence is increasing worldwide. Although the prognosis of patients with early- and low-grade EC is usually good, the outcomes of patients with advanced disease, deep muscle infiltration or metastasis and recurrence remain poor. The limited understanding of EC pathogenesis has hindered the development of effective diagnostic and therapeutic strategies. Based on the current WES of clinical EC patients with deletions or mutations in the FA gene, the present review identified a strong association between FA gene defects and EC, particularly high-grade EC (Table IV). The FA pathway is the core mechanism by which cells respond to inter-chain cross-linking (ICL) damage and maintain genomic stability and integrity (Fig. 1). In EC, defects in the FA gene lead to genomic instability and/or an abnormal cell cycle, thereby promoting tumorigenesis and progression (Fig. 2). Currently, research on the role of FA in EC primarily relies on clinical sample analysis and there is a lack of FA gene-deficient EC mouse models to validate the pathogenic mechanisms. In addition, the role of the FA gene in different EC subtypes and the associated mechanisms require further clarification. In this context, future studies should focus on establishing appropriate FA gene-deficient mouse models and refining EC subtypes to elucidate the complex pathogenesis of EC.

Table IV.

Molecular function of FA gene and molecular abnormalities associated with EC.

Table IV.

Molecular function of FA gene and molecular abnormalities associated with EC.

GeneAliasLocationMutation rate (%)Molecular functionsAbnormalities associated with EC development
FANCEFACE6p21.224.54Promotes DNA damage repair andLow expression (25)
genomic stability (21,29) andOverexpression (26)
involved in cell cycle regulation (25)Mutations (24)
FANCCFAC9q22.35.10Promotes DNA damage repair and genomic stability (24) and is involved in cell cycle regulation (24)Mutations (21,29)
FANCAFAA16q24.38.88Participates in DSB and ICL repair processes (35,36) and enhances the interaction between MSH2 and MLH1, participating in mismatch repair (37)Mutations (21,3841)
FANCD2FAD23p25.39.07Participates in the FA pathway byMutations (24)
interacting with FANCI to form an ID2 complex (42)Overexpression (9,44,45)
FANCI KIAA179415q26.18.70Forms ID2 complex (46) and regulatesSilencing (47)
FA core protein expression and the cell cycle (47)Mutations (4951)
FANCN PALB2/BRCA216p12.26.62Promotes the correct binding of RAD51 to DNA, participating in HR repair (54) and interacts with BRCA1/2 to form BRCA1-PALB2-BRCA2 axis, promoting DSB repair (55)Mutations (62,64,66,67)
FANCORAD51C17q223.21Promotes HR repair by interacting with other RAD51 homologs and is involved in cell cycle regulation (6870)Mutations (64,71)
FANCJ BACH1/BRIP117q23.27.37Binds to the carboxyl terminal of BRCA1 protein to promote HR repair (72) and promotes DNA end excision ICL and DSB (73,74)Mutations (60,71,77,78)
FANCD1BRCA213q13.115.31Binds to PALB2, initiates DNA end resection and promotes RAD51 loading, collaborates with BARD1 to enhanceMutations (77,8486)
RAD51 recombinase activity (80) and is involved in cell cycle regulation (80)
FANCSBRCA117q21.319.26Protects stalled DNA replication forks (81) and is involved in cell cycle regulation (80)Mutations (77,8486)
FANCRRAD5115q15.13.40Promotes homology search and double-Mutations (21)
stranded exchange by binding to single-Deletion (88)
stranded DNA to form nuclear protein filament (87)Polymorphism (8993)
FANCUXRCC27q36.13.02Involved in HR repair of ICLs andPolymorphism
DSBs (94,95)(100,101)
Mutations (98,99)
FANCWRFWD316q23.14.73Promotes the timely removal of RPA and RAD51 from DNA damage sites (102) and is involved in cell cycle regulation (80)Mutations (21)

[i] FA, Fanconi anemia; EC, endometrial cancer; DSB, DNA double-strand break; ICL, interstrand cross-linking; ID2, FANCI-FANCD2; HR, homologous recombination.

The present review described the pathogenesis of EC in terms of FA pathway abnormalities. An in-depth understanding of the molecular mechanisms of EC pathogenesis may facilitate the development of targeted therapies that specifically target key genes, leading to a more accurate treatment for EC, which may substantially improve the quality of life and survival of patients with high-grade and advanced EC. The present review provided new insights into the genetic factors and pathogenesis of EC and will facilitate the development of new EC treatments in the future.

Acknowledgements

Figure 1 was created using Figdraw (www.figdraw.com) and Figure 2 was generated using BioGDP (BioGDP.com).

Funding

The present study was supported by grants from Research Fund for Lin He's Academician Workstation of New Medicine and Clinical Translation in Jining, Medical University (grant no. JYHL2021MS13) and College Students' Innovation Training Program of Jining, Medical University (grant nos. cx2024138z, cx2024096z and cx2024007z).

Availability of data and materials

Not applicable.

Authors' contributions

Conceptualization was performed by LZ and AH, and methodology by MY and CL. Literature search and analysis were performed by MY, CL, HP, KM, XL, QL, YQ and ZZ. Investigation was performed by all authors. MY and CL were responsible for software and AH was responsible for supervision. Visualization was performed by MY and CL and writing-review and editing by LZ and AH. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EC

endometrial cancer

EAC

endometrioid adenocarcinoma

SCNA

somatic copy number alterations

MSI

microsatellite instability

FA

Fanconi anemia

ICL

interstrand cross-linking

DDR

DNA damage response

HR

homologous recombination

ID2

FANCI-FANCD2

TLS

trans lesion synthesis

DSBs

DNA double-strand breaks

LS

Lynch syndrome

UC

uterine cancer

MMR

mismatch repair

A-EMS

atypical endometriosis

TCGA

The Cancer Genome Atlas

WES

whole-exome sequencing

SNP

single nucleotide polymorphism

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI

2 

Feng J, Lin R, Li H, Wang J and He H: Global and regional trends in the incidence and mortality burden of endometrial cancer, 1990–2019: Updated results from the global burden of disease study, 2019. Chin Med J (Engl). 137:294–302. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Abdol Manap N, Ng BK, Phon SE, Abdul Karim AK, Lim PS and Fadhil M: Endometrial cancer in pre-menopausal women and younger: Risk factors and outcome. Int J Environ Res Public Health. 19:90592022. View Article : Google Scholar : PubMed/NCBI

4 

Gu B, Shang X, Yan M, Li X, Wang W, Wang Q and Zhang C: Variations in incidence and mortality rates of endometrial cancer at the global, regional, and national levels, 1990–2019. Gynecol Oncol. 161:573–580. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Bokhman JV: Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. 15:10–17. 1983. View Article : Google Scholar : PubMed/NCBI

6 

Bae HS, Kim H, Kwon SY, Kim KR, Song JY and Kim I: Should endometrial clear cell carcinoma be classified as type II endometrial carcinoma? Int J Gynecol Pathol. 34:74–84. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Suarez AA, Felix AS and Cohn DE: Bokhman Redux: Endometrial cancer ‘types’ in the 21st century. Gynecol Oncol. 144:243–249. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Amant F, Moerman P, Neven P, Timmerman D, Van Limbergen E and Vergote I: Endometrial cancer. Lancet. 366:491–505. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Mhawech-Fauceglia P, Wang D, Kim G, Sharifian M, Chen X, Liu Q, Lin YG, Liu S and Pejovic T: Expression of DNA repair proteins in endometrial cancer predicts disease outcome. Gynecol Oncol. 132:593–598. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Cancer Genome Atlas Research Network, . Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, Robertson AG, Pashtan I, Shen R, et al: Integrated genomic characterization of endometrial carcinoma. Nature. 497:67–73. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Matthews HK, Bertoli C and de Bruin RAM: Cell cycle control in cancer. Nat Rev Mol Cell Biol. 23:74–88. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Milletti G, Strocchio L, Pagliara D, Girardi K, Carta R, Mastronuzzi A, Locatelli F and Nazio F: Canonical and noncanonical roles of fanconi anemia proteins: Implications in cancer predisposition. Cancers (Basel). 12:26842020. View Article : Google Scholar : PubMed/NCBI

13 

Zhang X, Joseph S, Wu D, Bowser JL and Vaziri C: The DNA Damage Response (DDR) landscape of endometrial cancer defines discrete disease subtypes and reveals therapeutic opportunities. NAR Cancer. 6:zcae0152024. View Article : Google Scholar : PubMed/NCBI

14 

Nalepa G and Clapp DW: Fanconi anemia and the cell cycle: New perspectives on aneuploidy. F1000Prime Rep. 6:232014. View Article : Google Scholar : PubMed/NCBI

15 

García-de-Teresa B, Rodríguez A and Frias S: Chromosome instability in Fanconi anemia: From breaks to phenotypic consequences. Genes (Basel). 11:15282020. View Article : Google Scholar : PubMed/NCBI

16 

Wang W: Emergence of a DNA-damage response network consisting of Fanconi anaemia and BRCA proteins. Nat Rev Genet. 8:735–748. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Huang Y, Leung JWC, Lowery M, Matsushita N, Wang Y, Shen X, Huong D, Takata M, Chen J and Li L: Modularized functions of the Fanconi anemia core complex. Cell Rep. 7:1849–1857. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Ciccia A, Ling C, Coulthard R, Yan Z, Xue Y, Meetei AR, Laghmani el H, Joenje H, McDonald N, de Winter JP, et al: Identification of FAAP24, a Fanconi anemia core complex protein that interacts with FANCM. Mol Cell. 25:331–343. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Liang CC, Li Z, Lopez-Martinez D, Nicholson WV, Vénien-Bryan C and Cohn MA: The FANCD2-FANCI complex is recruited to DNA interstrand crosslinks before monoubiquitination of FANCD2. Nat Commun. 7:121242016. View Article : Google Scholar : PubMed/NCBI

20 

Kim H and D'Andrea AD: Regulation of DNA cross-link repair by the Fanconi anemia/BRCA pathway. Genes Dev. 26:1393–1408. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Salomão VMR, de Almeida AM, Matuo R and Sousa FG: In silico studies of molecular alterations in fanconi anemia genes from cancer cell lines and samples. Obs Econ Latinoam. 21:13067–13087. 2023.

22 

Pace P, Johnson M, Tan WM, Mosedale G, Sng C, Hoatlin M, de Winter J, Joenje H, Gergely F and Patel KJ: FANCE: The link between Fanconi anaemia complex assembly and activity. EMBO J. 21:3414–3423. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Bouffard F, Plourde K, Bélanger S, Ouellette G, Labrie Y and Durocher F: Analysis of a FANCE splice isoform in regard to DNA repair. J Mol Biol. 427:3056–3073. 2015. View Article : Google Scholar : PubMed/NCBI

24 

de Angelis de Carvalho N, Niitsuma BN, Kozak VN, Costa FD, de Macedo MP, Kupper BEC, Silva MLG, Formiga MN, Volc SM, Aguiar Junior S, et al: Clinical and molecular assessment of patients with Lynch syndrome and sarcomas underpinning the association with MSH2 germline pathogenic variants. Cancers (Basel). 12:18482020. View Article : Google Scholar : PubMed/NCBI

25 

Zheng C, Ren Z, Chen H, Yuan X, Suye S, Yin H and Fu C: Reduced FANCE confers genomic instability and malignant behavior by regulating cell cycle progression in endometrial cancer. J Cancer. 14:2670–2685. 2023. View Article : Google Scholar : PubMed/NCBI

26 

Zhou Z, Yin H, Suye S, He J and Fu C: Pan-cancer analysis of the prognostic and immunological role of Fanconi anemia complementation group E. Front Genet. 13:10249892023. View Article : Google Scholar : PubMed/NCBI

27 

Lin B, Li H, Zhang T, Ye X, Yang H and Shen Y: Comprehensive analysis of macrophage-related multigene signature in the tumor microenvironment of head and neck squamous cancer. Aging (Albany NY). 13:5718–5747. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Niedzwiedz W, Mosedale G, Johnson M, Ong CY, Pace P and Patel KJ: The Fanconi anaemia gene FANCC promotes homologous recombination and error-prone DNA repair. Mol Cell. 15:607–620. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Heald B, Mokhtary S, Nielsen SM, Rojahn S, Yang S, Michalski ST and Esplin ED: Unexpected actionable genetic variants revealed by multigene panel testing of patients with uterine cancer. Gynecol Oncol. 166:344–350. 2022. View Article : Google Scholar : PubMed/NCBI

30 

Luebben SW, Kawabata T, Johnson CS, O'sullivan MG and Shima N: A concomitant loss of dormant origins and FANCC exacerbates genome instability by impairing DNA replication fork progression. Nucleic Acids Res. 42:5605–5615. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Heinrich MC, Silvey KV, Stone S, Zigler AJ, Griffith DJ, Montalto M, Chai L, Zhi Y and Hoatlin ME: Posttranscriptional cell cycle-dependent regulation of human FANCC expression. Blood. 95:3970–3977. 2000. View Article : Google Scholar : PubMed/NCBI

32 

Gallmeier E, Calhoun ES, Rago C, Brody JR, Cunningham SC, Hucl T, Gorospe M, Kohli M, Lengauer C and Kern SE: Targeted disruption of FANCC and FANCG in human cancer provides a preclinical model for specific therapeutic options. Gastroenterology. 130:2145–2154. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Li X, Plett PA, Yang Y, Hong P, Freie B, Srour EF, Orschell CM, Clapp DW and Haneline LS: Fanconi anemia type C-deficient hematopoietic stem/progenitor cells exhibit aberrant cell cycle control. Blood. 102:2081–2084. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Singh DK, Gamboa RS, Singh AK, Walkemeier B, Van Leene J, De Jaeger G, Siddiqi I, Guerois R, Crismani W and Mercier R: The FANCC-FANCE-FANCF complex is evolutionarily conserved and regulates meiotic recombination. Nucleic Acids Res. 51:2516–2528. 2023. View Article : Google Scholar : PubMed/NCBI

35 

Ahsan MD, Webster EM, Qazi M, Weiss J, Levi S, Cantillo E, Chapman-Davis E, Holcomb K, Sharaf R and Frey MK: #1090 The mutational landscape of uterine sarcoma: Is there rationale for targeted therapies? Int J Gynecol Cancer. 33 (Suppl 3):A202.2–A202. 2023.

36 

Nasioudis D, Latif NA, Ko EM, Cory L, Kim SH, Martin L, Simpkins F and Giuntoli R II: Next generation sequencing reveals a high prevalence of pathogenic mutations in homologous recombination DNA damage repair genes among patients with uterine sarcoma. Gynecol Oncol. 177:14–19. 2023. View Article : Google Scholar : PubMed/NCBI

37 

Williams SA, Wilson JB, Clark AP, Mitson-Salazar A, Tomashevski A, Ananth S, Glazer PM, Semmes OJ, Bale AE, Jones NJ and Kupfer GM: Functional and physical interaction between the mismatch repair and FA-BRCA pathways. Hum Mol Genet. 20:4395–4410. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Drusbosky L, Haynes G, Grant B, Sobernis P and Lheureux S: Abstract A012: Genomic landscape of somatic alterations identified in endometrial cancer using liquid biopsy. Clin Cancer Res. 30 (5 Suppl):A0122024. View Article : Google Scholar

39 

Kral J, Jelinkova S, Zemankova P, Vocka M, Borecka M, Cerna L, Cerna M, Dostalek L, Duskova P, Foretova L, et al: Germline multigene panel testing of patients with endometrial cancer. Oncol Lett. 25:2162023. View Article : Google Scholar : PubMed/NCBI

40 

Liu Y, Gusev A and Kraft P: Germline cancer gene expression quantitative trait loci influence local and global tumor mutations. medRxiv. 2022.2008.2023.22279002. 2022.

41 

Wepy C, Nucci MR and Parra-Herran C: Atypical endometriosis: Comprehensive characterization of clinicopathologic, immunohistochemical, and molecular features. Int J Gynecol Pathol. 43:70–77. 2024. View Article : Google Scholar : PubMed/NCBI

42 

Lopez-Martinez D, Kupculak M, Yang D, Yoshikawa Y, Liang CC, Wu R, Gygi SP and Cohn MA: Phosphorylation of FANCD2 inhibits the FANCD2/FANCI complex and suppresses the Fanconi anemia pathway in the absence of DNA damage. Cell Rep. 27:2990–3005.e5. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Zheng C, Ren Z, Chen H, Yuan X, Suye S, Yin H, Zhou Z and Fu C: FANCD2 promotes the malignant behavior of endometrial cancer cells and its prognostic value. Exp Cell Res. 421:1133882022. View Article : Google Scholar : PubMed/NCBI

44 

Zhao Z, Wang R, Wang R, Song J, Ma F, Pan H, Gao C, Wang D, Chen X and Fan X: Pancancer analysis of the prognostic and immunological role of FANCD2: A potential target for carcinogenesis and survival. BMC Med Genomics. 17:692024. View Article : Google Scholar : PubMed/NCBI

45 

Lin HH, Zeng WH, Yang HK, Huang LS, Pan R and Lei NX: Fanconi anemia complementation group D2 promotes sensitivity of endometrial cancer cells to chemotherapeutic agents by inhibiting the ferroptosis pathway. BMC Womens Health. 24:412024. View Article : Google Scholar : PubMed/NCBI

46 

Ishiai M, Kitao H, Smogorzewska A, Tomida J, Kinomura A, Uchida E, Saberi A, Kinoshita E, Kinoshita-Kikuta E, Koike T, et al: FANCI phosphorylation functions as a molecular switch to turn on the Fanconi anemia pathway. Nat Struct Mol Biol. 15:1138–1146. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Kaljunen H, Taavitsainen S, Kaarijärvi R, Takala E, Paakinaho V, Nykter M, Bova GS and Ketola K: Fanconi anemia pathway regulation by FANCI in prostate cancer. Front Oncol. 13:12608262023. View Article : Google Scholar : PubMed/NCBI

48 

Zhao Y, Li Q, Li J, Cui Y and Lu Z: Expression and clinical significance of FANCI gene in pan-cancer: A comprehensive analysis based on multi-omics data. Front Genet. 16:15428882025. View Article : Google Scholar : PubMed/NCBI

49 

Fierheller CT, Alenezi WM, Serruya C, Revil T, Amuzu S, Bedard K, Subramanian DN, Fewings E, Bruce JP, Prokopec S, et al: Molecular genetic characteristics of FANCI, a proposed new ovarian cancer predisposing gene. Genes (Basel). 14:2772023. View Article : Google Scholar : PubMed/NCBI

50 

Wen H, Xu Q, Sheng X, Li H, Wang X and Wu X: Prevalence and landscape of pathogenic or likely pathogenic germline variants and their association with somatic phenotype in unselected Chinese patients with gynecologic cancers. JAMA Netw Open. 6:e23264372023. View Article : Google Scholar : PubMed/NCBI

51 

Dong L, Wang T, Li N, Yao H, Ying J, Wu L and Yuan G: Prevalence and prognostic relevance of homologous recombination repair gene mutations in uterine serous carcinoma. Cells. 11:35632022. View Article : Google Scholar : PubMed/NCBI

52 

Bi J, Areecheewakul S, Li Y, Yang S, Zhang Y, Ebeid K, Li L, Thiel KW, Zhang J, Dai D, et al: MTDH/AEG-1 downregulation using pristimerin-loaded nanoparticles inhibits Fanconi anemia proteins and increases sensitivity to platinum-based chemotherapy. Gynecol Oncol. 155:349–358. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Becker AE, Hernandez YG, Frucht H and Lucas AL: Pancreatic ductal adenocarcinoma: Risk factors, screening, and early detection. World J Gastroenterol. 20:11182–11198. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Buisson R and Masson JY: PALB2 self-interaction controls homologous recombination. Nucleic Acids Res. 40:10312–10323. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Foo TK and Xia B: BRCA1-dependent and independent recruitment of PALB2-BRCA2-RAD51 in the DNA damage response and cancer. Cancer Res. 82:3191–3197. 2022. View Article : Google Scholar : PubMed/NCBI

56 

Huang KL, Mashl RJ, Wu Y, Ritter DI, Wang J, Oh C, Paczkowska M, Reynolds S, Wyczalkowski MA, Oak N, et al: Pathogenic germline variants in 10,389 adult cancers. Cell. 173:355–370.e14. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Teo ZL, Park DJ, Provenzano E, Chatfield CA, Odefrey FA, Nguyen-Dumont T kConFab, Dowty JG, Hopper JL, Winship I, et al: Prevalence of PALB2 mutations in Australasian multiple-case breast cancer families. Breast Cancer Res. 15:R172013. View Article : Google Scholar : PubMed/NCBI

58 

Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, Vaccari EM, Bissonnette J, Booker JK, Cremona ML, et al: Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Genet Med. 18:823–832. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Fulk K, Milam MR, Li S, Yussuf A, Black MH, Chao EC, LaDuca H and Stany MP: Women with breast and uterine cancer are more likely to harbor germline mutations than women with breast or uterine cancer alone: A case for expanded gene testing. Gynecol Oncol. 152:612–617. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Heeke AL, Pishvaian MJ, Lynce F, Xiu J, Brody JR, Chen WJ, Baker TM, Marshall JL and Isaacs C: Prevalence of homologous recombination-related gene mutations across multiple cancer types. JCO Precis Oncol. 2018.PO.17.00286. 2018. View Article : Google Scholar : PubMed/NCBI

61 

Chan GHJ, Ong PY, Low JJH, Kong HL, Ow SGW, Tan DSP, Lim YW, Lim SE and Lee SC: Clinical genetic testing outcome with multi-gene panel in Asian patients with multiple primary cancers. Oncotarget. 9:306492018. View Article : Google Scholar : PubMed/NCBI

62 

Johnatty SE, Pesaran T, Dolinsky J, Yussuf A, LaDuca H, James PA, O'Mara TA and Spurdle AB: Case-case analysis addressing ascertainment bias for multigene panel testing implicates BRCA1 and PALB2 in endometrial cancer. Hum Mutat. 42:1265–1278. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Tian W, Bi R, Ren Y, He H, Shi S, Shan B, Yang W, Wang Q and Wang H: Screening for hereditary cancers in patients with endometrial cancer reveals a high frequency of germline mutations in cancer predisposition genes. Int J Cancer. 145:1290–1298. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Kondrashova O, Shamsani J, O'Mara TA, Newell F, McCart Reed AE, Lakhani SR, Kirk J, Pearson JV, Waddell N and Spurdle AB: Tumor signature analysis implicates hereditary cancer genes in endometrial cancer development. Cancers (Basel). 13:17622021. View Article : Google Scholar : PubMed/NCBI

65 

Cilento MA, Poplawski NK, Paramasivam S, Thomas DM and Kichenadasse G: Germline PALB2 variants and PARP inhibitors in endometrial cancer. J Natl Compr Canc Netw. 19:1212–1217. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Foo TK, Tischkowitz M, Simhadri S, Boshari T, Zayed N, Burke KA, Berman SH, Blecua P, Riaz N, Huo Y, et al: Compromised BRCA1-PALB2 interaction is associated with breast cancer risk. Oncogene. 36:4161–4170. 2017. View Article : Google Scholar : PubMed/NCBI

67 

Bowman-Colin C, Xia B, Bunting S, Klijn C, Drost R, Bouwman P, Fineman L, Chen X, Culhane AC, Cai H, et al: Palb2 synergizes with Trp53 to suppress mammary tumor formation in a model of inherited breast cancer. Proc Natl Acad Sci USA. 110:8632–8637. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Somyajit K, Subramanya S and Nagaraju G: Distinct roles of FANCO/RAD51C protein in DNA damage signaling and repair: Implications for Fanconi anemia and breast cancer susceptibility. J Biol Chem. 287:3366–3380. 2012. View Article : Google Scholar : PubMed/NCBI

69 

Kolinjivadi AM, Chong ST, Choudhary R, Sankar H, Chew EL, Yeo C, Chan SH and Ngeow J: Functional analysis of germline RAD51C missense variants highlight the role of RAD51C in replication fork protection. Hum Mol Genet. 32:1401–1409. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Chun J, Buechelmaier ES and Powell SN: Rad51 paralog complexes BCDX2 and CX3 act at different stages in the BRCA1-BRCA2-dependent homologous recombination pathway. Mol Cell Biol. 33:387–395. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Ring KL, Bruegl AS, Allen BA, Elkin EP, Singh N, Hartman AR, Daniels MS and Broaddus RR: Germline multi-gene hereditary cancer panel testing in an unselected endometrial cancer cohort. Mod Pathol. 29:1381–1389. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Hirshfield KM, Rebbeck TR and Levine AJ: Germline mutations and polymorphisms in the origins of cancers in women. J Oncol. 2010:2976712010. View Article : Google Scholar : PubMed/NCBI

73 

Nath S and Nagaraju G: FANCJ helicase promotes DNA end resection by facilitating CtIP recruitment to DNA double-strand breaks. PLoS Genet. 16:e10087012020. View Article : Google Scholar : PubMed/NCBI

74 

Alayev A, Salamon RS, Manna S, Schwartz NS, Berman AY and Holz MK: Estrogen induces RAD51C expression and localization to sites of DNA damage. Cell Cycle. 15:3230–3239. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Muhseena NK, Mathukkada S, Das SP and Laha S: The repair gene BACH1-a potential oncogene. Oncol Rev. 15:5192021. View Article : Google Scholar : PubMed/NCBI

76 

Long B, Lilyquist J, Weaver A, Hu C, Gnanaolivu R, Lee KY, Hart SN, Polley EC, Bakkum-Gamez JN, Couch FJ and Dowdy SC: Cancer susceptibility gene mutations in type I and II endometrial cancer. Gynecol Oncol. 152:20–25. 2019. View Article : Google Scholar : PubMed/NCBI

77 

de Jonge MM, Auguste A, van Wijk LM, Schouten PC, Meijers M, Ter Haar NT, Smit VTHBM, Nout RA, Glaire MA, Church DN, et al: Frequent homologous recombination deficiency in high-grade endometrial carcinomas. Clin Cancer Res. 25:1087–1097. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Nakamura K, Aimono E, Tanishima S, Imai M, Nagatsuma AK, Hayashi H, Yoshimura Y, Nakayama K, Kyo S and Nishihara H: Olaparib monotherapy for BRIP1-mutated high-grade serous endometrial cancer. JCO Precis Oncol. 4:PO.19.00368. 2020.

79 

Matsuzaki K, Borel V, Adelman CA, Schindler D and Boulton SJ: FANCJ suppresses microsatellite instability and lymphomagenesis independent of the Fanconi anemia pathway. Genes Dev. 29:2532–2546. 2015. View Article : Google Scholar : PubMed/NCBI

80 

Liu Y and Lu LY: BRCA1 and homologous recombination: Implications from mouse embryonic development. Cell Biosci. 10:492020. View Article : Google Scholar : PubMed/NCBI

81 

Kolinjivadi AM, Sannino V, de Antoni A, Técher H, Baldi G and Costanzo V: Moonlighting at replication forks-a new life for homologous recombination proteins BRCA1, BRCA2 and RAD51. FEBS Lett. 591:1083–1100. 2017. View Article : Google Scholar : PubMed/NCBI

82 

Deng CX: BRCA1: Cell cycle checkpoint, genetic instability, DNA damage response and cancer evolution. Nucleic Acids Res. 34:1416–1426. 2006. View Article : Google Scholar : PubMed/NCBI

83 

Lee H: Cycling with BRCA2 from DNA repair to mitosis. Exp Cell Res. 329:78–84. 2014. View Article : Google Scholar : PubMed/NCBI

84 

de Jonge MM, de Kroon CD, Jenner DJ, Oosting J, de Hullu JA, Mourits MJE, Gómez Garcia EB, Ausems MGEM, Margriet Collée J, van Engelen K, et al: Endometrial cancer risk in women with germline BRCA1 or BRCA2 mutations: Multicenter cohort study. J Natl Cancer Inst. 113:1203–1211. 2021. View Article : Google Scholar : PubMed/NCBI

85 

Jamieson A, Sobral de Barros J, Cochrane DR, Douglas JM, Shankar S, Lynch BJ, Leung S, Martin S, Senz J, Lum A, et al: Targeted and shallow whole-genome sequencing identifies therapeutic opportunities in p53abn endometrial cancers. Clin Cancer Res. 30:2461–2474. 2024. View Article : Google Scholar : PubMed/NCBI

86 

Smith ES, Da Cruz Paula A, Cadoo KA, Abu-Rustum NR, Pei X, Brown DN, Ferrando L, Sebastiao APM, Riaz N, Robson ME, et al: Endometrial cancers in BRCA1 or BRCA2 germline mutation carriers: Assessment of homologous recombination DNA repair defects. JCO Precis Oncol. 3:PO.19.00103. 2019.

87 

Ito M, Fujita Y and Shinohara A: Positive and negative regulators of RAD51/DMC1 in homologous recombination and DNA replication. DNA Repair (Amst). 134:1036132024. View Article : Google Scholar : PubMed/NCBI

88 

Auguste A, Genestie C, De Bruyn M, Adam J, Le Formal A, Drusch F, Pautier P, Crosbie EJ, MacKay H, Kitchener HC, et al: Refinement of high-risk endometrial cancer classification using DNA damage response biomarkers: A TransPORTEC initiative. Mod Pathol. 31:1851–1861. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Smolarz B and Romanowicz H: Association between single nucleotide polymorphism of DNA repair genes and endometrial cancer: A case-control study. Int J Clin Exp Pathol. 11:1732–1738. 2018.PubMed/NCBI

90 

Romanowicz-Makowska H, Smolarz B, Połać I and Sporny S: Single nucleotide polymorphisms of RAD51 G135C, XRCC2 Arg188His and XRCC3 Thr241Met homologous recombination repair genes and the risk of sporadic endometrial cancer in Polish women. J Obstet Gynaecol Res. 38:918–924. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Zeng X, Zhang Y, Yang L, Xu H, Zhang T, An R and Zhu K: Association between RAD51 135 G/C polymorphism and risk of 3 common gynecological cancers: A meta-analysis. Medicine (Baltimore). 97:e112512018. View Article : Google Scholar : PubMed/NCBI

92 

Michalska MM, Samulak D, Romanowicz H and Smolarz B: Association of polymorphisms in the 5′ untranslated region of RAD51 gene with risk of endometrial cancer in the Polish population. Arch Gynecol Obstet. 290:985–991. 2014. View Article : Google Scholar : PubMed/NCBI

93 

Krupa R, Sobczuk A, Popławski T, Wozniak K and Blasiak J: DNA damage and repair in endometrial cancer in correlation with the hOGG1 and RAD51 genes polymorphism. Mol Biol Rep. 38:1163–1170. 2011. View Article : Google Scholar : PubMed/NCBI

94 

Park JY, Virts EL, Jankowska A, Wiek C, Othman M, Chakraborty SC, Vance GH, Alkuraya FS, Hanenberg H and Andreassen PR: Complementation of hypersensitivity to DNA interstrand crosslinking agents demonstrates that XRCC2 is a Fanconi anaemia gene. J Med Genet. 53:672–680. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Johnson RD, Liu N and Jasin M: Mammalian XRCC2 promotes the repair of DNA double-strand breaks by homologous recombination. Nature. 401:397–399. 1999. View Article : Google Scholar : PubMed/NCBI

96 

Griffin CS, Simpson PJ, Wilson CR and Thacker J: Mammalian recombination-repair genes XRCC2 and XRCC3 promote correct chromosome segregation. Nat Cell Biol. 2:757–761. 2000. View Article : Google Scholar : PubMed/NCBI

97 

Webster ALH, Sanders MA, Patel K, Dietrich R, Noonan RJ, Lach FP, White RR, Goldfarb A, Hadi K, Edwards ME, et al: Fanconi anemia pathway deficiency drives copy number variation in squamous cell carcinomas. bioRXiv. 2021.2008. 2014.456365. 2021.PubMed/NCBI

98 

Nero C, Pasciuto T, Cappuccio S, Corrado G, Pelligra S, Zannoni GF, Santoro A, Piermattei A, Minucci A, Lorusso D, et al: Further refining 2020 ESGO/ESTRO/ESP molecular risk classes in patients with early-stage endometrial cancer: A propensity score-matched analysis. Cancer. 128:2898–2907. 2022. View Article : Google Scholar : PubMed/NCBI

99 

Taylor NP, Gibb RK, Powell MA, Mutch DG, Huettner PC and Goodfellow PJ: Defective DNA mismatch repair and XRCC2 mutation in uterine carcinosarcomas. Gynecol Oncol. 100:107–110. 2006. View Article : Google Scholar : PubMed/NCBI

100 

Michalska MM, Samulak D, Bieńkiewicz J, Romanowicz H and Smolarz B: Association between-41657C/T single nucleotide polymorphism of DNA repair gene XRCC2 and endometrial cancer risk in Polish women. Pol J Pathol. 66:67–71. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Romanowicz H, Brys M, Forma E and Smolarz B: Lack of association between the 4234G/C X-ray repair cross-complementing 2 (XRCC2) gene polymorphism and the risk of endometrial cancer among Polish population. J Gynecol Res Obstet. 2:47–50. 2016. View Article : Google Scholar

102 

Inano S, Sato K, Katsuki Y, Kobayashi W, Tanaka H, Nakajima K, Nakada S, Miyoshi H, Knies K, Takaori-Kondo A, et al: RFWD3-mediated ubiquitination promotes timely removal of both RPA and RAD51 from DNA damage sites to facilitate homologous recombination. Mol Cell. 66:622–634.e8. 2017. View Article : Google Scholar : PubMed/NCBI

103 

Fu X, Yucer N, Liu S, Li M, Yi P, Mu JJ, Yang T, Chu J, Jung SY, O'Malley BW, et al: RFWD3-Mdm2 ubiquitin ligase complex positively regulates p53 stability in response to DNA damage. Proc Natl Acad Sci USA. 107:4579–4584. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Wang Y: Role of WD domain-containing proteins in cell cycle progression (unpublished thesis). University of Illinois at Urbana-Champaign; 2017

Related Articles

Journal Cover

November-2025
Volume 32 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yao M, Liu C, Ping H, Meng K, Li X, Li Q, Qi Y, Zhu Z, Zhang L, Han A, Han A, et al: Potential role of Fanconi anemia pathway in the pathogenesis of endometrial cancer (Review). Mol Med Rep 32: 295, 2025.
APA
Yao, M., Liu, C., Ping, H., Meng, K., Li, X., Li, Q. ... Han, A. (2025). Potential role of Fanconi anemia pathway in the pathogenesis of endometrial cancer (Review). Molecular Medicine Reports, 32, 295. https://doi.org/10.3892/mmr.2025.13660
MLA
Yao, M., Liu, C., Ping, H., Meng, K., Li, X., Li, Q., Qi, Y., Zhu, Z., Zhang, L., Han, A."Potential role of Fanconi anemia pathway in the pathogenesis of endometrial cancer (Review)". Molecular Medicine Reports 32.5 (2025): 295.
Chicago
Yao, M., Liu, C., Ping, H., Meng, K., Li, X., Li, Q., Qi, Y., Zhu, Z., Zhang, L., Han, A."Potential role of Fanconi anemia pathway in the pathogenesis of endometrial cancer (Review)". Molecular Medicine Reports 32, no. 5 (2025): 295. https://doi.org/10.3892/mmr.2025.13660