Abstract Oxaliplatin, a key chemotherapeutic agent, often induces resistance in colorectal cancer (CRC) treatment, highlighting the urgent need for reliable biomarkers to predict treatment efficacy. In this study, we aimed to identify key genes associated with oxaliplatin resistance in CRC and to evaluate their potential as prognostic biomarkers. Using CRC patient data from the TCGA dataset, we categorized patients into oxaliplatin-resistant and -sensitive groups and conducted differential expression analysis. Key feature genes were identified through univariate Cox analysis, LASSO regression, and stepwise multivariate Cox regression. The predictive value of the identified markers was validated using logistic regression, weighted gene co-expression network analysis (WGCNA), and external validation in GEO cohorts. The tumor microenvironment (TME) was assessed using the MCP-counter algorithm, and CRC cell experiments were performed to evaluate changes in drug sensitivity following oxaliplatin exposure. Based on TCGA CRC data, we constructed a prognostic index derived from a three-gene signature associated with oxaliplatin resistance. This index was significantly correlated with progression-free survival (PFS) in oxaliplatin-resistant CRC patients and showed robust prognostic performance, with AUCs of 0.848 and 0.861 in gastric cancer and pancreatic adenocarcinoma cohorts, respectively. Notably, TNFAIP2 knockout significantly reduced clonogenic ability in CRC cells following oxaliplatin treatment. Our results identify TLE4, TNFAIP2, and ARGLU1 as key contributors to oxaliplatin resistance in CRC. The oxaliplatin resistance–related gene signature (ORGSig) serves as a promising tool for predicting treatment response and prognosis in CRC patients receiving oxaliplatin-based chemotherapy. This signature also offers potential for guiding personalized therapy and overcoming drug resistance in clinical practice. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-05556-2. Keywords: Chemoresistance, Oxaliplatin, CRC, Prognostic marker, TNFAIP2 Subject terms: Cancer, Cell biology, Gastroenterology Introduction Colorectal cancer (CRC) is among the most prevalent malignancies of the digestive system, with over one million new cases diagnosed annually^[32]1. Current treatment strategies include surgical resection, radiotherapy, chemotherapy, targeted therapy, and immunotherapy^[33]2–[34]4. Oxaliplatin, a key chemotherapeutic agent, is widely used in combination with 5-fluorouracil and folinic acid for CRC treatment^[35]5. Although oxaliplatin has improved clinical outcomes, approximately 40–50% of patients with stage II or III CRC develop resistance, resulting in poor prognosis^[36]6. Therefore, identifying biomarkers associated with oxaliplatin resistance and prognosis is crucial for addressing CRC’s molecular heterogeneity and improving therapeutic outcomes. Oxaliplatin, a third-generation platinum-based chemotherapeutic agent, inhibits tumor progression by disrupting DNA synthesis and impairing cell division in cancer cells^[37]7–[38]9. Its role in CRC treatment was first established in 1996^[39]10. Although oxaliplatin can be used as monotherapy, it is commonly administered in combination with other agents to enhance efficacy. In advanced and metastatic CRC, it is typically combined with 5-fluorouracil and folinic acid. Beyond CRC, oxaliplatin also exhibits antitumor activity in breast, bladder, prostate, brain, and lung cancers, as well as lymphomas^[40]11–[41]15. Despite its clinical benefits, resistance frequently emerges, especially in aggressive tumors. Mechanisms contributing to chemoresistance include enhanced DNA repair, inhibition of apoptosis, altered drug transport, detoxification, and epigenetic modifications^[42]16. Recent studies have shown that UPF1 upregulation promotes oxaliplatin resistance by regulating TOP2A activity and maintaining cancer stemness in CRC^[43]17. Additionally, long non-coding RNAs (lncRNAs) and feedback loops contribute significantly to the regulation of chemotherapeutic responses in tumor cells^[44]18. These findings underscore the complexity of oxaliplatin resistance mechanisms. However, the molecular heterogeneity of CRC hinders a clear understanding of these mechanisms, highlighting the need to further explore the genes involved. Developing a gene panel based on oxaliplatin resistance–related genes is therefore essential for enhancing CRC treatment and prognosis. In this study, we focused on three key genes—TLE4, TNFAIP2, and ARGLU1—which were significantly upregulated in oxaliplatin-resistant CRC samples. These genes were selected for their established roles in cell proliferation, tumor immunity, and chemoresistance. TLE4 is associated with tumor progression and poor prognosis in CRC; TNFAIP2 is involved in inflammatory signaling and drug resistance; and ARGLU1 plays a central role in DNA damage repair across multiple cancer types. Together, these genes form a predictive panel that may help stratify CRC patients based on their susceptibility to oxaliplatin resistance. Our objective was to improve personalized prognosis prediction and treatment selection for CRC patients receiving oxaliplatin-based chemotherapy. To this end, we developed an oxaliplatin resistance–related gene signature (ORGSig) that demonstrates strong predictive power for both prognosis and treatment response. We also experimentally validated the functional role of TNFAIP2 in CRC cells, providing further insight into the mechanisms driving tumor progression and chemoresistance in CRC. Results DEGs between oxaliplatin-resistant and oxaliplatin-sensitive patients in TCGA dataset To identify key genes associated with oxaliplatin resistance in CRC, we selected patients from the TCGA-CRC cohort who had received oxaliplatin-based chemotherapy. Among the 64 patients with complete chemotherapy data, 50 were classified as sensitive and 14 as resistant. Compared to the sensitive group, the resistant group showed upregulation of 397 genes and downregulation of 69 genes (Fig. [45]1A, B; Table [46]S1). To explore the biological functions of these differentially expressed genes (DEGs), we performed Gene Ontology (GO) and pathway enrichment analyses using Metascape. The upregulated genes were significantly enriched in: (1) immune response and cell activation pathways (e.g., inflammatory response, leukocyte migration, cell activation, organismal homeostasis); (2) biological regulation processes (e.g., epithelial cell proliferation, population-level proliferation, phosphorus metabolism regulation, cell adhesion); and (3) signaling and interaction pathways (e.g., integrin-mediated cell surface interactions) (Fig. [47]1C). These 397 upregulated genes were considered candidate components for constructing the ORGSig. Fig. 1. [48]Fig. 1 [49]Open in a new tab DEGs between oxaliplatin-resistant and oxaliplatin-sensitive patients in TCGA dataset. (A, B) Volcano plot and heatmap illustrating the DEGs between the chemosensitive group and chemoresistant group in the TCGA cohort. (C) A total of 397 upregulated genes were subjected to pathway enrichment analysis using the Metascape platform. Construction of ORGSig for CRC To identify key genes with the greatest prognostic impact in CRC patients treated with oxaliplatin, we performed univariate Cox regression analysis on the 397 upregulated genes. This analysis identified 32 genes significantly associated with poor prognosis (Fig. [50]2A). LASSO regression further narrowed the candidates to four genes: TLE4, TNFAIP2, CRABP2, and ARGLU1 (Fig. [51]2B, C). Subsequent stepwise multivariate Cox regression analysis refined the selection to three critical genes—TLE4, TNFAIP2, and ARGLU1—for constructing the prognostic model. Fig. 2. [52]Fig. 2 [53]Open in a new tab Construction of ORGSig for CRC. (A) 32 ORGSig showing an adverse prognosis correlation in the TCGA cohort. (B) The tuning parameter (λ) for LASSO regression was determined via 10-fold cross-validation. (C) Changes in LASSO coefficients and alternative ORGSig across different values of the tuning parameter (λ). (D) ROC curves demonstrating the performance of the three-gene panel in CRC samples from the training set. Verification of ORGSig for CRC To evaluate the predictive performance of the ORGSig model for chemotherapy response, we conducted ROC curve analysis on 114 CRC patients with available chemotherapy records, yielding an AUC of 0.788 (Fig. [54]2D). To further validate the robustness of ORGSig, we tested the model in both the TCGA training cohort and an external validation cohort ([55]GSE87211). In the TCGA cohort, stratification based on risk scores demonstrated significantly improved progression-free survival (PFS) in the low-risk group compared to the high-risk group, thereby confirming the prognostic utility of the model (Fig. [56]3A). ROC analysis showed AUCs of 0.791, 0.767, and 0.937 for 1-, 3-, and 5-year PFS predictions, respectively (Fig. [57]3B), indicating strong temporal predictive accuracy. For external validation, we selected oxaliplatin-treated samples from [58]GSE87211. Patients in the high-risk group exhibited significantly poorer survival (P < 0.05; Fig. [59]3C). The corresponding ROC analysis yielded AUCs of 0.669, 0.579, and 0.666 for 1-, 3-, and 7-year survival predictions, respectively (Fig. [60]3D). Collectively, these results support the prognostic reliability and predictive utility of ORGSig in oxaliplatin-treated CRC patients. Fig. 3. [61]Fig. 3 [62]Open in a new tab Verification of ORGSig for CRC. (A) The Kaplan–Meier survival curves showing notable variations in PFS based on different risk scores in the TCGA cohort. (B) Time-dependent ROC curve analysis for 1-, 3-, and 7-year survival in the TCGA cohort. (C) The Kaplan–Meier survival curves showing notable variations in PFS based on different risk scores in the GEO cohort. (D) Time-dependent ROC curve analysis for 1-, 3-, and 7-year survival in the GEO cohort. The application of the prognostic nomogram To determine whether ORGSig serves as an independent prognostic factor, we performed univariate and multivariate Cox regression analyses. In the univariate analysis, M stage, clinical stage, and risk score were significantly associated with prognosis (Fig. [63]4A). Multivariate analysis further confirmed that the risk score remained an independent predictor, with a hazard ratio (HR) of 1.460 (95% CI: 1.240–1.700, P < 0.001) (Fig. [64]4B). To enhance the clinical utility of ORGSig, we constructed a nomogram integrating the risk score with key clinicopathological variables to predict 1-, 2-, and 3-year PFS (Fig. [65]4C). As expected, higher risk scores were associated with poorer outcomes. Calibration plots showed strong concordance between predicted and observed survival probabilities, indicating good model reliability (Fig. [66]4D). In addition, ROC curve analyses demonstrated that the risk score outperformed traditional clinical parameters (T, N, and M stages), with AUC values of 0.763, 0.713, and 0.741 at different time points (Fig. [67]4E). Collectively, these findings support ORGSig as a robust and clinically valuable prognostic tool for CRC patients. Fig. 4. [68]Fig. 4 [69]Open in a new tab The application of the prognostic nomogram. (A, B) Univariate and multivariate Cox regression analyses evaluating association between clinicopathological factors and ORGSig with PFS.(C) Nomogram for predicting PFS. (D) The calibration curves for 1-,3- and 5- year PFS. (E) The ROC curve of risk score and clinical characteristics was performed based on 1-, 3-, and 5-year PFS. Three genes as the best combination in predicting tumor progression To further validate the reliability of ORGSig, we performed multivariable logistic regression to assess its ability to predict tumor progression after chemoresistance. This analysis provided deeper insight into the association between the gene panel and disease progression. We constructed multiple regression models using individual genes and combined gene sets to compare their AUC values (Fig. [70]5A). Among all combinations, the model incorporating all three core biomarkers achieved the highest AUC of 0.797, indicating superior predictive performance (Fig. [71]5B). While the AUCs of other combinations were slightly lower, they still demonstrated considerable predictive potential (Fig. [72]5C). These findings support the hypothesis that the three key genes contribute to chemoresistance and are associated with differences in PFS in CRC. To evaluate the generalizability of ORGSig, we extended the analysis to other oxaliplatin-treated cancers in the TCGA database, including stomach adenocarcinoma (STAD, n = 22) and pancreatic adenocarcinoma (PAAD, n = 20). Multivariable logistic regression was applied to each cancer type, and AUC values were calculated for different gene combinations. In both STAD and PAAD cohorts, the three-gene model showed the highest predictive accuracy, with AUCs of 0.848 and 0.861, respectively (Fig. [73]5D–I). Fig. 5. [74]Fig. 5 [75]Open in a new tab Three genes as the best combination in predicting tumor progression. (A, D, G) Evaluation of all gene combinations and their corresponding AUC values calculated by a multivariate logistic regression model in patients with COREAD, STAD, and PAAD. (B, C, E, F, H, I) ROC curves depicting the performance of the top two combinations in predicting clinical outcomes for patients with COREAD, STAD, and PAAD. Applying WCGNA to ORGSig and clinical characteristics To further validate the reliability of ORGSig in predicting tumor progression after CRC chemotherapy, we applied Weighted Gene Co-expression Network Analysis (WGCNA). This approach enabled us to investigate associations among ORGSig, PFS, risk score, and gene modules with co-expression patterns similar to those of the key genes. We selected a soft-thresholding power of β = 8 to construct a scale-free network (Fig. [76]6A). By merging modules with similar expression profiles using dynamic tree cutting, we obtained a consolidated network comprising seven modules (Fig. [77]6B). The relationships between each module and clinical outcomes, including PFS and risk score, are shown in Fig. [78]6C. Notably, PFS showed a negative correlation with risk score, highlighting the model’s effectiveness in predicting chemoresistance and tumor recurrence. The three core genes were located in distinct modules: TLE4 in the brown module (risk score correlation: 0.5; PFS correlation: − 0.02), TNFAIP2 in the yellow module (risk score: 0.43; PFS: − 0.04), and ARGLU1 in the turquoise module (risk score: 0.36; PFS: 0.01). Gene enrichment analysis further revealed that these modules were enriched in pathways related to post-chemotherapy cell proliferation, metabolism, and immune responses (Fig. [79]6D–F). Fig. 6. [80]Fig. 6 [81]Open in a new tab Applying WCGNA to ORGSig and clinical characteristics. (A) Determination of the soft threshold for WGCNA (power = 8) based on scale independence and mean connectivity. (B) Construction of a cluster tree using all genes from CRC patients treated with oxaliplatin, with similar genes grouped into color-coded modules, and merged dynamically. (C) Heatmap matrix displaying the relationship between gene modules and clinical traits. (D-F) Dot chart illustrating pathways enriched through GO analysis. TLE4, TNFAIP2 and ARGLU1 were classified into brown, yellow and turquoise modules respectively. Associations of ORGSig model with immune cell infiltration Based on the enrichment analysis of DEGs, which revealed activation of immune-related pathways, we further explored immunological differences between high- and low-risk score groups. Using the ESTIMATE algorithm, we calculated immune, stromal, and ESTIMATE scores in the TCGA cohort. The high-risk group exhibited significantly higher immune infiltration and stromal content, as reflected by elevated immune, stromal, and ESTIMATE scores (Fig. [82]7A). MCP-counter analysis revealed increased infiltration of multiple immune cell types—including cytotoxic T cells, NK cells, B cells, monocytes, myeloid dendritic cells, endothelial cells, and cancer-associated fibroblasts—in the high-risk group (Fig. [83]7B). To investigate potential immunotherapeutic targets, we examined immune checkpoint gene expression and found that HHLA2 was more highly expressed in the low-risk group, whereas TNFRSF9, TIGIT, CD28, CD80, HAVCR2, CD200R1, BTLA, CD276, NRP1, CTCN1, ICOS, IDO1, CTLA4, LAG3, and TNFRSF8 were significantly upregulated in the high-risk group (Fig. [84]7C). Additionally, we assessed treatment sensitivity and observed that high-risk patients showed better responses to anti-CTLA4 therapy and chemotherapy (P < 0.05, Fig. [85]7D). Collectively, these findings suggest that the high-risk score group exhibits enhanced immune activity and may benefit from immunotherapy, providing important insights for personalized treatment strategies. Fig. 7. [86]Fig. 7 [87]Open in a new tab Associations of ORGSig model with immune cell infiltration. (A) Histogram depicting immune cell infiltration analysis.(B) Comparison of immune cell proportions based on MCP counter between the low-risk and high-risk groups in the TCGA cohort. (C) Expression distribution of common immune checkpoints in high- and low-risk score patients from the TCGA cohort. (D) Sensitivity analysis for anti-PD-1 and anti-CTLA4 treatments in high- and low-risk groups. TNFAIP2 confers OXA resistance in CRC cells in vitro The role of TNFAIP2 in CRC initiation, progression, and chemoresistance remains incompletely understood. In this study, we focused on its potential involvement in oxaliplatin resistance. Data from the Human Protein Atlas revealed elevated TNFAIP2 expression in CRC tissues (Fig. [88]8A). Immunohistochemistry (IHC) analysis further confirmed significantly higher TNFAIP2 levels in the oxaliplatin-resistant group compared to the sensitive group (Fig. [89]8B, C). To evaluate the prognostic significance of TNFAIP2 in CRC, we utilized the Kaplan–Meier Plotter online tool. High TNFAIP2 expression was significantly associated with poor overall survival (OS) (P = 0.01, Fig. [90]8D). Subgroup analysis indicated a trend toward reduced survival in patients undergoing surgery alone (P = 0.14, Fig. [91]8E) and a significant association with poor prognosis in patients receiving postoperative chemotherapy (P = 0.019, Fig. [92]8F). To validate the functional role of TNFAIP2 in oxaliplatin resistance, we established TNFAIP2 knockdown SW480 cell lines using shRNA, with silencing efficiency confirmed by qRT-PCR (Fig. [93]8G). Colony formation assays showed a significant reduction in clonogenic potential following TNFAIP2 silencing under baseline conditions. Upon oxaliplatin treatment, knockdown cells exhibited a near-complete loss of colony-forming ability (Fig. [94]8H, I). Additionally, TNFAIP2 knockdown markedly enhanced oxaliplatin-induced apoptosis compared to control cells (Fig. [95]8J, K). Collectively, these results suggest that TNFAIP2 plays a key role in mediating oxaliplatin resistance in CRC and may serve as a promising therapeutic target to overcome chemoresistance. Fig. 8. [96]Fig. 8 [97]Open in a new tab TNFAIP2 confers OXA resistance in CRC cells in vitro. (A) Immunohistochemical data of TNFAIP2 in HPA database. Scale bar = 100 μm. (B, C) Representative IHC images of TNFAIP2 in 8 CRC tissues with different groups. Scale bar = 100 μm. (D, E, F) Kaplan–Meier survival curves of CRC patients with TNFAIP2 expression in the Kaplan–Meier Plotter database. (G) qRT–PCR was employed to validate the knockdown efficiency in various cell lines. (H, I) Analysis of the clonogenic assay. (J, K) Annexin V-FITC and PI staining showing apoptosis in the indicated CRC cells treated with OXA (30 μM) for 48 h. Discussion Oxaliplatin is widely used in the treatment of CRC, but resistance to this agent remains a significant clinical challenge^[98]19. Tumor cells that acquire resistance can evade oxaliplatin-induced cytotoxicity and contribute to tumor recurrence, particularly in relapsed and metastatic settings^[99]20–[100]22. Previous studies have demonstrated a strong association between dysregulated gene expression and chemotherapy resistance^[101]23. Consequently, there is increasing interest in identifying reliable prognostic biomarkers to predict response to oxaliplatin-based chemotherapy and guide treatment strategies. Compared with previous studies, our work was distinguished by a rigorous, multi-step strategy to identify candidate genes associated with oxaliplatin resistance. For example, Yin et al. developed a gene signature based on resistance-related pathways and validated it across multiple cohorts. In contrast, we employed a comprehensive pipeline that incorporated differential expression analysis, univariate Cox regression, LASSO regression, and multivariate Cox regression, ensuring both statistical robustness and clinical relevance^[102]24. Unlike approaches that rely on single datasets or specific pathways, we utilized TCGA data for gene identification and further validated our findings using the GEO database and other independent tumor cohorts, thereby enhancing generalizability. While Zhou et al. focused on lncRNAs linked to oxaliplatin sensitivity, our study targets protein-coding genes directly implicated in resistance mechanisms, offering clearer therapeutic implications^[103]25. Furthermore, although Lin et al. proposed a resistance-related signature based on large-scale colon cancer profiling, our model integrates gene expression data with survival outcomes and external validation, yielding a robust and clinically applicable prognostic tool^[104]26. This integrative approach improves predictive accuracy and supports the development of personalized treatment strategies for CRC patients receiving oxaliplatin. To explore the mechanisms underlying oxaliplatin resistance, we analyzed CRC samples from the TCGA database by comparing oxaliplatin-resistant and sensitive groups. Differential expression analysis identified a set of upregulated genes, which were subsequently subjected to enrichment analysis. These genes showed significant associations with tumor immunity, cell signaling, and malignant biological processes. While both upregulated and downregulated genes can contribute to drug resistance, our study focused on upregulated genes for several reasons. First, upregulated genes are often actively involved in cellular pathways that mediate resistance, facilitating tumor cells’ ability to evade the cytotoxic effects of oxaliplatin. Second, their elevated expression may reflect an adaptive response to chemotherapy, highlighting their potential as biomarkers or therapeutic targets. In contrast, downregulated genes typically represent loss of normal cellular functions, which are less directly associated with resistance mechanisms. We therefore prioritized upregulated genes to identify those most functionally relevant to oxaliplatin resistance. Using a stepwise analytical pipeline, we first identified DEGs, followed by univariate Cox regression to evaluate associations with patient survival. The resulting candidates were refined using LASSO and multivariate Cox regression, yielding three key genes: TLE4, TNFAIP2, and ARGLU1 (Fig. [105]2). These genes were not only significantly upregulated in resistant samples but also correlated with poor prognosis, enhancing their clinical relevance. The predictive performance of this three-gene signature was subsequently validated using independent CRC cohorts from the GEO database, as well as other oxaliplatin-treated tumor types from the TCGA database. These results confirm the robustness and translational potential of the identified gene panel. TLE4 is a member of the highly conserved TLE transcriptional corepressor family. While TLE proteins do not directly bind DNA, they interact with transcription factors such as Hes1 and Runx2 to form repressive complexes^[106]27. Previous research suggests TLE4 functions as a tumor suppressor in hematologic malignancies and is located on chromosome 9q, a region frequently deleted in acute myeloid leukemia (AML)^[107]28. Loss of TLE4 expression promotes leukemic cell proliferation and activates the Wnt signaling pathway, enhancing pro-inflammatory signaling^[108]29. In contrast, elevated TLE4 expression in CRC correlates with advanced Dukes stage, lymph node metastasis, and poor prognosis, likely through activation of the JNK–c-Jun pathway, which promotes proliferation and invasion^[109]30. Additionally, TLE4 polymorphisms have been linked to shorter PFS following gemcitabine treatment in HER2-negative metastatic breast cancer, indicating a role in chemoresistance^[110]31. Wei et al. also reported TLE4 overexpression in cisplatin-resistant bladder cancer xenografts, supporting its broader involvement in drug resistance^[111]32. Together, these findings suggest TLE4 may suppress tumor development in hematologic cancers but contribute to chemoresistance in solid tumors such as CRC and bladder cancer. Further studies are warranted to clarify TLE4’s role in oxaliplatin resistance and evaluate its potential as a therapeutic target or predictive biomarker in CRC. ARGLU1 is a well-conserved bifunctional protein that serves as both an activator and effector in the nuclear receptor system. Its overexpression inhibits the proliferation of gastric and head and neck squamous cell carcinoma cells, whereas silencing ARGLU1 suppresses the growth and migration of breast cancer cells^[112]33–[113]35. Functionally, ARGLU1 plays a key role in transcriptional regulation and the DNA damage response. While its overexpression promotes cancer cell proliferation, its downregulation produces the opposite effect. Notably, ARGLU1 also contributes to chemoresistance by enhancing DNA repair capacity^[114]36. These findings are consistent with our data, which show that reducing ARGLU1 expression sensitizes CRC cells to oxaliplatin. TNFAIP2 is a novel oncoprotein implicated in the initiation and progression of multiple cancers^[115]37–[116]39. Elevated TNFAIP2 expression has been identified as a risk factor in cervical cancer, contributing to increased susceptibility and incidence^[117]40. In gastric cancer, its serum levels are elevated and associated with inflammation and tumor angiogenesis^[118]41. TNFAIP2 overexpression is also linked to cisplatin resistance by promoting epithelial–mesenchymal transition (EMT) in urothelial carcinoma cells^[119]42. In head and neck squamous cell carcinoma, high TNFAIP2 expression correlates with poor prognosis and chemoresistance. Mechanistically, TNFAIP2 competes with KEAP1 to stabilize NRF2, thereby inhibiting cisplatin-induced oxidative stress and apoptosis^[120]43. In breast cancer, TNFAIP2 promotes chemoresistance by activating RAC1 via IQGAP1^[121]44. Collectively, these findings support a strong role for TNFAIP2 in mediating resistance to platinum-based therapies. Our results further confirm its high expression in oxaliplatin-resistant CRC tissues and its association with poor prognosis. Silencing TNFAIP2 in CRC cells enhances sensitivity to oxaliplatin, underscoring its potential as a therapeutic target. Furthermore, this study primarily focuses on patients receiving oxaliplatin-based chemotherapy. Future research should explore the potential interactions between oxaliplatin and other therapeutic modalities, such as immune checkpoint inhibitors, to gain deeper insight into the mechanisms and clinical implications of oxaliplatin resistance in CRC. Comprehensive investigation into combination treatment strategies is essential to better understand their effects on resistance development and to refine predictive models for improved clinical applicability. Conclusions In conclusion, this study identified three key genes—TLE4, TNFAIP2, and ARGLU1—associated with oxaliplatin resistance in CRC patients from the TCGA-CRC dataset. The resulting three-gene signature demonstrated strong prognostic value and clinical relevance, offering a novel and effective tool for predicting oxaliplatin resistance and guiding personalized treatment strategies. Furthermore, the model exhibited potential applicability across other cancer types, underscoring its broader translational significance. Future studies should aim to validate this gene panel in larger, prospective clinical cohorts and work toward developing standardized protocols to facilitate its integration into clinical practice. Methods and material Data collection Clinical and transcriptomic data for the TCGA CRC cohort were obtained from the TCGA repository in TPM-normalized format for downstream analyses. After excluding samples lacking prognostic information, 114 cases were included in the prognostic evaluation and designated as the training cohort, among which 64 had documented chemotherapy response data. For external validation of the prognostic model, the [122]GSE87211 dataset from the GEO database was used as an independent validation cohort. A total of 83 cases receiving oxaliplatin-based chemotherapy were selected from 243 available samples based on treatment records. Identification of differentially expressed ORGSig To identify pivotal genes associated with chemotherapy, a cohort of 64 cases with comprehensive chemotherapy records from the TCGA dataset was chosen. Chemotherapy responses in all 64 cases were characterized into four categories: complete response (CR), partial response (PR), stable disease (SD), and clinical progressive disease (PD). For analytical clarity, patients exhibiting SD and PD were grouped as chemoresistant, while those displaying CR and PR were categorized as chemosensitive in terms of chemotherapy response. Differential expression analysis of ORGSig between these two groups was conducted using the “limma” package, employing criteria of P < 0.05 and |log2(Foldchange[FC])| > mean (|Log2(FC)|) + 2 SD (|Log2(FC)|)^[123]45. Development and validation of prognostic models in both the training and validation cohorts A prognostic model was established using the TCGA dataset as the training cohort. Initially, univariate cox proportional hazard regression analysis was implemented to identify DEGs with significant prognostic implications (P < 0.01). Subsequently, the LASSO technique combined with cox proportional hazards regression was employed to reduce variables and prevent overfitting. Lastly, multivariate stepwise cox regression analysis was performed to select the risk model comprising the ORGSig, defining the resultant model as the ORGSig. Risk score for each sample was calculated as follows: Risk Score = 0.7047583×TLE4[exp] + 0.4238049×TNFAIP2[exp] + 0.4774622×ARGLU1[exp]. For validation of the prognostic model, patients in the training cohort were stratified into low- and high-risk score groups based on the median risk score. The divergence in PFS between these groups was evaluated employing Kaplan–Meier survival analysis. Time-dependent ROC curve was constructed to determine the AUC for 1-year, 3-year, and 5-year PFS in CRC patients. This analysis was replicated in the external validation dataset [124]GSE87211. Association between risk score and immune characteristics The MCP-counter algorithm was utilized to determine the enriched presence of immune cells, endothelial cells, and fibroblasts in individual samples^[125]46. Prediction of response to immune checkpoint blockade therapy relied on the TIDE model^[126]47. Additionally, the expression levels of conventional immune checkpoints were compared in different groups. Development and evaluation of the nomogram in the TCGA dataset To delve deeper into identifying independent prognostic factors among various potential variables, univariate and multivariate analyses using the cox regression model were performed. Subsequently, a nomogram integrating the ORGSig and independent prognostic clinical factors was designed for practical applicability. Testing the applicability of ORGSig In addition to TCGA-CRC, we extended our analysis to include patients who underwent oxaliplatin treatment from various TCGA projects to validate the effectiveness of the Oxaliplatin resistance gene panel. Logistic regression models were constructed using the R package “glmnet” showcasing various gene combinations and their corresponding models. The top-performing models and their respective AUC values for distinct cancer types were presented accordingly. WGCNA analysis The R package “WGCNA” was employed to establish a co-expression network. The methodology involved selecting TCGA-CRC patients who received oxaliplatin, integrating all mRNA into a gene expression cluster analysis. Following sample clustering and removal of outliers, the soft-thresholding power was determined using an algorithm to create a scale-free co-expression network and identify gene modules. Each module, distinguished by a distinct color, encompassed genes demonstrating analogous expression profiles. Through dynamic tree cut analysis, genes were classified, and modules sharing high similarity were merged to form cohesive dynamic clusters. Furthermore, the relationship between different modules and two clinical attributes, PFS and risk score, was examined. Modules containing crucial Oxaliplatin-resistant genes were prioritized for GO analysis. Patient sample A total of 16 fresh tissue samples were obtained from patients, including 8 cases of oxaliplatin-sensitive and 8 cases of oxaliplatin-resistant colorectal cancer patients (stages II-III) who received adjuvant chemotherapy based on oxaliplatin after surgery. All tissues were embedded in paraffin to investigate the correlation between TNFAIP2 and oxaliplatin resistance. Tumor responses were evaluated using the Response Evaluation Criteria in Solid Tumors (RECIST). Written informed consent for the use of tumor tissues and clinical data in this study was obtained from all participants. This study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of Yancheng First Hospital. Cell cultures Immortalized human colorectal tumor cell lines SW480 was bought from cells bank of the Chinese Academy of Science (Shanghai, China). All cells were seeded in Dulbecco’s Modifed Eagle’s Medium (DMEM, Invitrogen, USA). 10% Fetal Bovine Serum (Clark bioscience, USA) and ampicillin and streptomycin (Gibco, USA) were added to the DMEM. All cells were cultured in 5% CO[2] at 37 ℃. Cellular transfection The shRNA transfections were performed by the manufacturer’s protocol. shTNFAIP2#1 (CTTCACCAAAGGGAAGAAGAA), shTNFAIP2#2 (CCACAAACTTCGTGGATCAAA) and shNC (UUUGUACUACACAAAAGUACUG) were purchased from GenePharma (Shanghai, China). IHC IHC was performed following a previously described method^[127]48. The following antibody was used: anti-TNFAIP2 (Santa Cruz Biotechnology, sc-28318; dilution 1:200). The staining intensity was evaluated manually by two independent, experienced pathologists using the following scoring system: 0 = no staining, 1 = weak staining, 2 = moderate staining, and 3 = strong staining. The percentage of positively stained cells was assessed using four categories: 1 (0–10%), 2 (11–50%), 3 (51–80%), and 4 (81–100%). The final IHC score was calculated by multiplying the intensity score by the percentage of positive cells. RNA isolation and qRT–PCR RNA isolation and qRT–PCR were conducted according to our previously described study^[128]48. The gene-specific primers are as follows (5’–3’): TNFAIP2-F, CCTGGGCCTGAAAGTTCCTT, R, 5’- GGCATCCTTTATACCGGCCA-3’. β-actin—F, ACCCTGAAGTACCCCATCGAG, R, AGCACAGCCTGGATAGCAAC. Colony formation assay In the colony formation assay, 1000 cells were plated in 6-well plates and allowed to grow for approximately 10 days with or without oxaliplatin treatment (2.0 µM)^[129]49. Subsequently, the colonies were stained with crystal violet and counted at the conclusion of the experiment. Flow cytometry analysis The transfected cells were cultured with 0 or 30 µM oxaliplatin for 48 h. Flow cytometry was performed using an Annexin V-FITC Apoptosis Detection Kit from Vazyme Biotech Co. Ltd. (Nanjing, China). Statistical analysis The data are expressed as means ± standard deviation (SD), with all experiments replicated in triplicate. Statistical analyses were conducted using GraphPad Prism 8.0 or R software (version 4.2.1). A significance threshold of P < 0.05 was utilized. Statistical significance is denoted as follows: ns, not significant; *P < 0.05; **P < 0.01; ***P < 0.001. Electronic supplementary material Below is the link to the electronic supplementary material. [130]Supplementary Material 1^ (38.5KB, xlsx) Acknowledgements