Breast cancer (BC) and endometrial cancer (EC) are two common malignant tumors in women often arising from "unresistant" estrogen stimulation. Endocrine therapy, comprising both estrogen receptor inhibitors and aromatase inhibitors (AI), is widely administered to patients with hormone receptor-positive BC worldwide. Tamoxifen (TAM) reduces the risk of BC recurrence and contralateral BC, but there are some side effects, primarily its proliferative impact on the endometrium, leading to EC[15]. Studies indicate that, 5-year adjuvant TAM therapy increased the risk of EC by 2.4 times compared with patients without adjuvant therapy[16, 17]. Even after adjusting for confounding factors (including age, BMI, diabetes, hypertension, dyslipidemia, PCOS, and GnRH agonist therapy), the use of TAM after BC still increased the risk of EC by about 4 times. Additionally, the risk of EC rises with prolonged TAM therapy duration. Patients who used TAM for 10 years face a 1.5%-3.2% increased risk compared to 5-year users[18]. However, it has been reported that a persistently high EC incidence in HR-negative BC patients who did not use TAM[19, 20]. A genome-based analysis found that EC patients reveals no significant differences in EC occurrence between TAM-exposed and non-exposed patients[11]. Furthermore, EC patients with a prior BC history were characterized by higher grades and more type II cancers than estrogen-dependent type I cancers[19, 21, 22], implying factors beyond TAM contribute, possibly stemming from shared genetic backgrounds or mutation characteristics.
In our study, we used TCGA and ICGC genome expression data to establish 11 gene prognostic features of BC-related EC patients, aiming to discover promising biomarkers and therapeutic targets for EC prognosis and treatment. Initially, differentially expressed BCRGs were identified in EC through differential analysis and WGCNA. Subsequently, the analyses of GO enrichment and KEGG pathway revealed their association with processes, including mitosis, nuclear division, chromosome segregation, and cell cycle regulation. DEGs were mainly enriched associated with the cell cycle, motor proteins, oocyte meiosis, and p53 signaling, suggesting their potential therapeutic implications in tumor division. 11 key genes were identified through survival analysis, LASSO, and Cox analysis, including ATAD2, CDKN2A, E2F1, GGH, MTHFD2, NPR1, NR3C1, PAMAR1, SRPX, STXBP1, and TTK. These genes showed significant associations with EC patient overall survival rates based on Kaplan-Meier analysis. ATAD2 is recognized as a gene responsive to estrogen and androgen in hormone-dependent cancer cells, functioning as a transcriptional co-regulator of ER and AR[23, 24]. In EC, its expression significantly increases compared to normal endometrium and precancerous lesions, and this elevation correlates with a poor prognosis[25]. TAD2 overexpression is closely associated with upregulation of genes linked to increased proliferation, indicating its role as either an upstream mediator or a co-factor in proliferation regulation. Similar regulatory effects are also observed in breast cancer[26]. CDKN2A functions as a negative regulator of the cell cycle by inhibiting cyclin-dependent kinases[27, 28]. Hypermethylation of CDKN2A is a common epigenetic aberration in various cancers, including BC and EC[29]. The E2F1 transcription factor family orchestrates the transition from G1 to S phase in the cell cycle by activating target genes through transcription[30, 31], playing a pivotal role cell proliferation regulation[32, 33]. Studies have demonstrated its direct correlation with unfavorable prognosis in breast cancer[34], ovarian cancer[35], and other cancer types. Studies have shown upregulation of E2F1 in EC tissues[36], and the mechanism may potentially linked to signaling pathways such as CDK4/RB/E2Fs axis[37]. Cell metabolism is upregulated in cancer cells to support tumor growth and metastasis, and GGH and MTHFD2 are both enzymes involved in folate metabolism. GGH's dysregulation affects DNA methylation and gene expression, impacting crucial biological pathways such as cell cycle regulation, development, and proliferation[38]. Elevated GGH expression has been observed in invasive breast cancer[39], ERG-negative prostate cancer[39] and colon cancer[40], compared with adjacent non-cancerous tissues, correlating with unfavorable prognoses and clinical outcomes. Additionally, Heightened expression of GGH can diminish the sensitivity of cancer cells to chemotherapeutic agents such as 5-fluorouracil, methotrexate, pemetrexed, and carboplatin[41, 42]. The one-carbon folate cycle is pivotal in cancer metabolism, facilitating the synthesis of nucleotides and amino acids that are essential for rapid cellular proliferation[43]. The one-carbon metabolic enzyme MTHFD2, crucial for nucleic acid synthesis, is encoded by the nucleus and operates within the mitochondria[44, 45]. While widely upregulated during embryogenesis, its expression in normal adult tissues is generally low or absent. MTHFD2 is markedly overexpressed in various tumors, and its expression level is associated with the progression of malignant tumors. Proposed mechanisms include P53 inactivation[46], oxidative stress[47, 48], anti-inflammatory immunity[49], and DNA damage repair[50, 51]. E Extensively studied in breast cancer, MTHFD2 is closely linked to its occurrence and poor prognosis[52-54]. its potential molecular mechanisms in EC remain unclear. Neuropilin-1 (NRP1), a transmembrane protein widely expressed in cancer cells, plays crucial roles in tumor proliferation, migration, and invasion[55, 56] by stimulating multiple growth factor receptors such as VEGF and EGF[55, 57]. NRP1 also modulates immune cell function within the tumor microenvironment, affecting the host's response to cancer[58]. Its expression in EC correlates with invasion ability and tumor grade[59], highlighting its potential as both a diagnostic and therapeutic target[60]. NR3C1, a key regulator of glucocorticoid action, is involved in several cellular processes, including tumor proliferation and differentiation, particularly when bound to glucocorticoids[61]. Extensive research has demonstrated that the upregulation of NR3C1 promotes tumor proliferation, metastasis, and drug resistance, such as triple-negative breast cancer, ovarian cancer[62, 63]. PAMR1, which contains a peptidase domain that contributes to muscle regeneration, was initially identified as downregulated in the muscles from mice with Duchenne muscular dystrophy (DMD). Recognized as a muscle protease vital for regeneration, PAMR1 is predominantly expressed in various tissues, including normal skeletal muscle and brain[64, 65]. In human malignant tumors, PAMR1 expression correlates with favorable cervical cancer prognoses[66]. PAMR1 has been shown to inhibit the growth of breast cancer cells and is frequently absent in breast cancer samples (20.8%-58.3%)[67, 68] . This absence has led to its classification as a potential tumor suppressor in breast cancer, often being suppressed due to promoter hypermethylation in breast cancer tissues[69]. However, the role of PAMAR1 in the occurrence and progression of EC remains unexplored. SRPX , a transmembrane protein composed of 464 amino acids and 3 sushi domains, was originally identified as a pathogenic gene in patients with X-linked retinoic syndrome[70]. It functions as a tumor suppressor gene with pro-apoptotic function, and has been observed to be downregulated in various human tumor cells and tissues[71] . Mouse gene knockout studies have further demonstrated its pro-apoptotic function[72]. Srpx knockout mice exhibit tumor development in around 30% of cases, encompassing lymphoma, lung cancer, and liver cancer[73]. Synapse binding protein 1 (STXBP1) primarily regulates the fusion of intracellular granular membranes and various exocytosis processes[74], i including vesicle fusion, initiation, docking, and membrane fusion. This protein is vital in eliminating tumor cells and facilitating granular cell membrane fusion[75]. Although exocytosis being prevalent in both normal and tumor cells, the expression and function of STXBP1 in tumor progression have been scarcely investigated. In lung adenocarcinoma, upregulated STXBP1 expression correlates with poor prognosis[76]. Threonine and tyrosine kinase (TTK), alternatively referred to as unipolar spindle 1 (Mps1), acts as a spindle assembly checkpoint, guaranteeing the precise segregation of chromosomes into daughter cells[77]. Apart from the testes and placenta, TTK is rarely detected in normal tissues[78]. However, high levels of TTK have been observed in various human malignancies, including breast cancer[79], ovarian cancer[80], pancreatic cancer[81] correlating with poor prognosis. Knockdown of TTK expression or treatment with TTK inhibitors can inhibit tumor growth by suppressing cell proliferation and invasion, leading to significant survival benefits[82-84]. In ovarian cancer, downregulation of TTK can sensitize cisplatin-resistant cells to cisplatin therapy[85], potentially positioning TTK as a therapeutic target in cancer treatment. Studies indicate that TTK expression is increased in EC, associated with poor prognosis, positively correlated with high TNM stage, and involved in immune infiltration. This suggests its promising role as a diagnostic biomarker for distinguishing EC tissue from normal tissue[86]. Additionally, TTK inhibitor (NTRC0066-0) has demonstrated significant inhibition of EC cells growth[87]. These findings underscore the close association of identified key genes with tumor occurrence and progression, primarily through cell cycle regulation, which is consistent with GO results. Nevertheless, additional research is necessary to clarify their specific mechanisms in modulating EC.
Combining biomarkers in the risk model can improve predictive capacity and expedite the formulation of personalized treatment strategies, surpassing the efficacy of single clinical biomarkers. BCRGRS, constructed from these 11 genes, emerged as a significant independent prognostic indicator for OS. Utilizing both TCGA and ICGC datasets, BCRGRS categorized patients into HRG and LRG by median value. LRG patients exhibited higher survival rates, whereas those in the HRG group had poorer survival outcomes. Specifically, the median OS of patients in the LRG group significantly exceeded that of patients in the HRG group. Univariate and multivariate Cox regression analyses confirmed BCRGRS as an independent prognostic factor. Although the survival curves in the ICGC cohort potentially intersect at a 30% risk of death, this does not imply superior survival rates for 30% of patients in the LRG group compared to those in the HRG group. At the intersection, 6 patients in the LRG group were still alive, whereas only 4 remained in the HRG group. Thus, a 30% risk of death does not equate to 30% of patients deceased. As the last patient in the LRG group succumbed, the LRG curve dropped to 0, unavoidably intersecting with the HRG curve. Extension of follow-up duration, with surviving patients in the LRG group, might preclude curve intersection. Additionally, our data revealed higher BCRGs scores in EC patients aged over 65 years, obese individuals, and those with the increase of EC FIGO stage and tumor grade, BCRGs scores were also higher. A nomogram combining age, stage, grade, and BCRGRS to validate its strong ability for predict EC prognosis, i suggesting the potential utility of BCRGRS as a clinical biomarker.
The tumor microenvironment (TME) plays a significant role in the occurrence, progression, and metastasis of malignant tumors. Comprising tumor cells, stromal cells, endothelial cells, immune cells, and extracellular matrix components produced by tumor-associated cells, the TME modulates immune responses, facilitating immune evasion and fostering tumor cell tolerance, ultimately impacting tumor pathogenesis. Identifying dependable prognostic indicators and immunotherapy targets is imperative in elucidating the dynamics of EC. Compared with other immune systems, the endometrial immune system exhibits distinctive characteristics, mainly manifested in two aspects: it prevents infection and defends against various bacteria and viruses, while also facilitating allogeneic embryo transplantation[88]. Upon comparing the immune cell infiltration and activation pathways between the LRG and HRG groups, we observed a general decrease in the infiltration of immune cells and a decrease in the activity of immune-related pathways in the HRG group. Extensive studies have demonstrated that substantial T cell infiltration, particularly by cytotoxic CD8 T cells, is associated with a favorable prognosis. CD8 T cells can eliminate tumor cells through the release of cytotoxic molecules such as granzyme and perforin[89, 90], and secrete IFN-γ to induce tumor ferroptosis[91], which holds prognostic value for EC[92, 93]. Furthermore, BCRGRS shows promise in guiding immunotherapy selection for EC patients. Studies indicates that the approval of immune checkpoint inhibitors, such as dostarlimab or pembrolizumab (PD-1 inhibitors), benefits approximately 20-30% of 20-30% of patients with advanced EC[94]. Our study revealed that BCRGRS and several immune checkpoint genes, including PD-L1 and CTLA-4, showed higher expression in the LRG group, suggesting potential benefits from anti-PD-1 and anti-CTLA-4 therapy for patients belonging to this group. TIDE predicts the response to immune checkpoint inhibitors (ICIs) by simulating two primary mechanisms of tumor immune evasion: the induction of T cell dysfunction in tumors with high cytotoxic T lymphocyte (CTL) infiltration, and the prevention of T cell infiltration in tumors with low CTL levels[95]. A positive correlation exists between the TIDE score and the possibility of tumor immune evasion, indicating that patients with higher TIDE scores may not benefit from ICI therapy. In our study, the LRG group exhibited lower TIDE scores and higher T cell dysfunction scores, whereas the HRG group displayed higher TIDE scores and increased T cell rejection scores. Consequently, HRG patients are more prone to T cell dysfunction and rejection when receiving immunotherapy, possibly leading to immune evasion as a consequence of T cell rejection. These patients typically exhibit a poor response to ICIs. In contrast, patients may benefit more from ICIs treatment. Furthermore, analysis of four different Immune Phenotype Scores (IPS) in the TCIA database revealed higher sensitivity in the LRG group, indicating that these patients might derive greater benefits from PD-1 and CTLA-4 inhibitors. External validation datasets, including urothelial carcinoma patients treated with anti-PD-L1 and malignant melanoma patients treated with anti-PD-1/CTLA-4, further substantiated the effectiveness of BCRGRS in predicting immunotherapy responses. In both immunotherapy cohorts, HRG patients exhibited lower OS and a higher incidence of disease progression following immunotherapy. This evidence suggests that BCRGRS can be considered an effective biomarker to predict response to immunotherapy.
n 2013, the TCGA Alliance initially categorized the prognosis of EC into four categories: POLE hypermutation, microsatellite instability hypermutation (MSIH), low copy number (CNL), and high copy number (CNH)[96]. Gene-based detection is increasingly pivotal in EC treatment, dividing the prognosis into various risk categories and guiding surgical and adjuvant therapies. In our analysis of gene mutations across various BCRGRS subgroups, missense variants emerged as the most prevalent, followed by nonsense variants and frameshift deletions. TP53 and PIK3CA mutations occurred more frequently in HRG, while PTEN, PIK3, and ARIDI mutations were more prevalent in LRG. TP53 mutations, the most common genetic events in cancer, are associated with malignant tumor invasion and poor prognosis[97, 98]. It affects the cancer cell cycle via the p53/TGFß signaling pathway. PIK3CA may promote tumor proliferation through the PI3K-AKT signaling pathway[99]. Consequently, HRG patients, who typically exhibit high levels of TP53 and PIK3CA mutations, face a worse prognosis compared to LRG patients with lower frequencies of these mutations, which is consistent with our findings. Some studies have demonstrated that tumor mutational burden (TMB) can reflect tumor neoantigen potential and is closely linked to DNA repair defects, predicting immunotherapy efficacy across various tumors. Patients with mismatch repair defects (dMMR) and high microsatellite instability (MSI-H) typically exhibit higher TMB[99, 100]. Treatment with immune checkpoint inhibitors often leads to improved response and survival benefits in patients with high TMB[101, 102]. Our study observed that BCRGRS scores were negatively correlated with TMB, with MSI-H patients having the lowest scores, aligning with our immune correlation analysis results. This suggests that LRG patients with EC combined with TMB-H and MSI-H may have a greater chance of responding positively to immunotherapy. Based on molecular typing system assessment, although there is no significant difference in POLE hypermutation scores and the three molecular subtypes, the proportion of POLE hypermutation with the best prognosis is higher than that of HRG, while the opposite is true for CNH with the worst prognosis. could function as an adjunctive tool for molecular typing in immunotherapy and as predictive markers, enhancing the accurate evaluation of EC patients' immune microenvironment and treatment effects.
The main limitations of our study were as follows. Firstly, due to data limitations, our study cohorts are sourced from different public datasets, inevitably leading to intra-tumor or intra-patient tumor heterogeneity. Secondly, owing to time constraints, although we identified the survival impact of related genes shared between EC and BC in EC patients, there has been no experimental validation or clinical trials conducted to date. The underlying mechanisms behind these phenomena remain unclear, necessitating further experimental evidence. Thirdly, the robustness of the prognostic model still needs verification and explanation through large-scale prospective studies and functional mechanism experiments. Currently, more work needs to be done to identify common biomarkers focusing on endometrial cancer and breast cancer.