International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Colon cancer remains a leading cause of cancer-related mortality worldwide, accounting for nearly 10% of all cancer deaths (1). While localized tumors exhibit favorable outcomes with >90% 5-year survival rates, metastatic disease continues to portend a dismal prognosis, with median survival under 30 months despite advances in systemic therapy (2). This stark contrast underscores the critical need for robust prognostic biomarkers and novel therapeutic strategies to improve patient stratification and treatment outcomes.
Recent breakthroughs in tumor immunology have reshaped our understanding of colon cancer progression. The tumor immune microenvironment, specifically the infiltration density and spatial organization of CD8+ cytotoxic T lymphocytes and CD45RO+ memory T cells, represents an independent prognostic factor beyond conventional TNM staging (3-5). Conversely, FOXP3+ regulatory T cells (Tregs) contribute to immunosuppression and correlate with tumor recurrence (6,7). However, despite these advances, current immune score systems remain incomplete, failing to incorporate emerging players such as CEACAM6, a glycoprotein increasingly implicated in immune evasion and metastasis (8-10).
Simultaneously, CEACAM6 (carcinoembryonic antigen-related cell adhesion molecule 6) has emerged as a key player in colon cancer progression (11,12). Originally identified as a marker of poor differentiation, CEACAM6 is now recognized to promote metastasis through multiple mechanisms including resistance to anoikis, enhancement of epithelial-mesenchymal transition and modulation of tumor-stroma interactions (13,14). Most recently, CEACAM6 has been implicated in immune evasion by upregulating PD-L1 expression and recruiting myeloid-derived suppressor cells (MDSCs) (15). These findings suggest that CEACAM6 may serve as both a prognostic marker and a potential therapeutic target in colon cancer.
A multicenter, dual-platform investigation was conducted, across three tertiary hospitals in China, analyzing 301 patients with colon cancer. Notably, the present study represents a significant expansion beyond our previous single-center pilot investigation (16), which analyzed 120 patients from one institution. The current multicenter design, larger sample size (301 vs. 120), incorporation of long-term overall survival (OS) data (follow-up until 2025), and multivariate Cox regression analysis which established CEACAM6 and FOXP3 as independent prognostic factors, collectively provide a higher level of evidence and move beyond initial correlation to establish prognostic value. Furthermore, the samples used herein are entirely new and non-overlapping with the previous publication, having been collected between 2015 and 2020 from three distinct medical centers. The present study investigated the associations of tumor-infiltrating lymphocytes (TILs), FOXP3+ regulatory T cells, and CEACAM6 expression with clinicopathological characteristics and prognosis in colon cancer, while exploring their roles in tumor progression and providing a theoretical foundation for immunotherapy development.
A total of 301 paraffin-embedded colon cancer tissue samples were collected from July 2015 to June 2020. The sample inclusion period was limited to June 2020 to ensure a minimum of 5 years of follow-up data for OS analysis, with the final follow-up date being June 30, 2025. This approach guaranteed the availability of complete and robust survival data for meaningful Kaplan-Meier and Cox regression analyses. The specimens for the present study were obtained through a collaborative effort among three independent tertiary hospitals. A total of 161 cases were collected from the Affiliated Hospital of Jiangnan University (Wuxi, China), while Jiangsu Provincial Veterans Hospital (Wuxi, China) and the First Affiliated Hospital of Soochow University (Suzhou, China) contributed 30 and 110 cases, respectively. It should be noted that these institutions share no direct administrative affiliation; their collaboration was solely facilitated by academic connections within our research team. To ensure consistency across all samples, each hospital collected specimens from their own patient archives using identical predefined inclusion and exclusion criteria. Prior to specimen acquisition, written informed consent was obtained from all participants for the use of their tissues in scientific research. Patients with prior exposure to radiotherapy, chemotherapy, or immunotherapeutic interventions were excluded from the study cohort. Histopathological examination confirmed the diagnosis of colon adenocarcinoma in all cases, with tumor staging (TNM classification) and histological grading performed in strict accordance with the Union for International Cancer Control guidelines (8th edition) (17,18). The present retrospective multicenter study was conducted with approval from the research Ethics Committee of all participating hospitals in accordance with the Declaration of Helsinki.
Sections (4-µΜ) were prepared from formalin-fixed, paraffin-embedded (FFPE) tissue blocks and subjected to standard deparaffinization through three xylene washes (5 min each) followed by graded ethanol rehydration (100, 95, 75 and 50%; 5 min each). Antigen retrieval was performed by heat-induced epitope retrieval in 10 mmol/l sodium citrate buffer (pH 6.0) at 100˚C for 15 min, after which endogenous peroxidase activity was blocked with 3% H2O2 in methanol (10 min, 25˚C). Non-specific binding was minimized by 10 min blocking with 10% normal horse serum (Wuhan Boster Biological Technology) at room temperature, followed by 2 h incubation in a humidified dark chamber with the following antihuman antibodies: Monoclonal mouse IgG against CD3 (cat. no. sc-20047), polyclonal rabbit IgG against CD4 (cat. no. sc-7219) and CD8 (cat. no. sc7188), (all from Santa Cruz Biotechnology, Inc.; 1:100); monoclonal mouse IgG2a against CD45RO (cat. no. ab86080; Abcam; 1:10), monoclonal rabbit IgG against CEACAM6 (cat. no. ab134074; Abcam; 1:400) and monoclonal mouse IgG3 against FOXP3 (cat. no. ab450; Abcam; 1:50). After PBS washing, sections were incubated with biotinylated polyclonal goat antimouse/rabbit IgG secondary antibodies (cat. no. K5007; Dako; Agilent Technologies, Inc.; 1:2,000) for 1 h under identical conditions, then developed with 3,3'-diaminobenzidine tetrahydrochloride hydrate and counterstained with hematoxylin (5 min) at room temperature. Five random high-power fields per section were analyzed by light microscopy (BX53; Olympus Corporation), excluding necrotic areas, with human tonsillar FFPE sections as positive controls (obtained with donor consent) and PBS substitution as negative controls, all procedures being performed in accordance with institutional biosafety protocols and manufacturer specifications.
IHC expression of CD3, CD4, CD8, CD45RO, CEACAM6 and FOXP3 was evaluated using a validated two-tiered scoring system incorporating both staining intensity and cellular distribution (19). All slides underwent blinded evaluation by two independent pathologists. The staining intensity was scored as 0 (achromatic), 1 (light yellow), 2 (brownish yellow) or 3 (brown). In addition, the percentage of positive cells was scored as 0 (<5%), 1 (5-25%), 2 (26-50%), 3 (51-75%), or 4 (>75%). The two scores were added together, and the samples were assigned to one of four levels as follows: (-), score 0-1; (+), score 2; (++), score 3-4; or (+++), score ≥5. () and (+) were defined as negative expression, (++) as weak expression and (+++) as strong expression. Discrepant scores between the two pathologists were resolved through joint re-evaluation until a consensus was reached.
Total RNA was extracted from FFPE tissue sections using the RNeasy FFPE Kit (cat. no. 73504; Qiagen, Inc.). Briefly, freshly cut sections were deparaffinized using the manufacturer's proprietary solution, followed by tissue lysis in an optimized buffer to release nucleic acids. To reverse formalin-induced cross-linking, samples were heated at 80˚C, then treated with DNase to eliminate genomic DNA contamination. Lysates were mixed with Buffer RBC, and ethanol was added to facilitate RNA binding to RNeasy MinElute spin columns. Purified RNA was eluted in a minimum of 14 µl of RNase-free water. First-strand cDNA was synthesized from total RNA using the RevertAid™ First Strand cDNA Synthesis kit (cat. no. K1622; Thermo Fisher Scientific, Inc.) following the manufacturer's protocol. qPCR was performed with FastStart Universal SYBR Green Master (Rox) kit (cat. no. 4913914001; Sigma-Aldrich; Merck KGaA) under the following conditions: Initial denaturation at 95˚C for 1 min; 45 cycles of 95˚C for 20 sec, 58˚C for 30 sec, and 68˚C for 45 sec. β-actin served as the endogenous control. Relative gene expression was calculated via the 2-ΔΔCq method using Cq values normalized to β-actin (20). All primers were synthesized by Sangon Biotech Co., Ltd., as shown in Table I.
All statistical analyses were carried out using SPSS (v26; IBM Corporation). Normality of continuous variables was assessed using Shapiro-Wilk tests. Normally distributed continuous variables are presented as the mean ± standard deviation. For comparisons of mRNA expression levels between groups, the independent samples t-test was used after confirming normality (Shapiro-Wilk test, P>0.05) and homogeneity of variances (Levene's test, P>0.05). The χ2 test was employed to assess the association between expression levels and patient characteristics. Survival was analyzed by Kaplan-Meier curves, and differences were assessed using the logrank test. Variables were selected by univariate and multivariate Cox regression analyses, with forest plots visualized using R (version 4.5.1). P<0.05 was considered to indicate a statistically significant difference.
From July 2015 to June 2020, a total of 301 patients with colon cancer were enrolled. The study population comprised 141 patients (46.8%) with early-stage disease (stage I-II) and 160 patients (53.2%) with advanced-stage disease (stage III-IV). Sex distribution showed a male predominance (n=174, 57.8%) compared with female patients (n=127, 42.2%). Tumor laterality was distributed with 159 cases (52.8%) presenting as left-sided tumors and 142 cases (47.2%) as right-sided tumors. Histopathological examination revealed poorly differentiated tumors in 96 cases (31.9%), while the majority exhibited well-to-moderately differentiated features (n=205, 68.1%). The mean age at the time of surgery was 64.94±10.63 years (range: 32-93 years).
IHC analysis of colon cancer specimens revealed distinct spatial distribution patterns of TILs, with infiltration densities significantly associated with tumor stage and differentiation (Table II). Early-stage tumors (I-II) exhibited significantly higher infiltration of CD3+ (P<0.001), CD8+ (P=0.001), and CD45RO+ T-cells (P<0.001) compared with advanced-stage (III-IV) tumors, whereas CD4+ T-cell infiltration showed an inverse correlation with tumor progression (P=0.014). Similarly, well-to-moderately differentiated tumors displayed significantly greater infiltration of CD3+ (P<0.001) and CD45RO+ T-cells (P<0.001) than poorly differentiated tumors (Fig. 1), while CD4+ T-cell density was reduced in higher-grade tumors (P=0.020). By contrast, no significant associations were found between TIL subsets and patient age (all P>0.05) or tumor location (P>0.05). Notably, sex differences were observed only for CD45RO+ T-cells (P=0.017), with female patients exhibiting higher infiltration levels than males.
Table IIRelationship between clinicopathological parameters and CD3+, CD4+, CD8+ and CD45RO+ T-cell infiltrations in patients with colon cancer. |
IHC staining demonstrated strong FOXP3 and CEACAM6 expression in 65.4% (119/182) and 61.8% (115/186) of cases, respectively (Table III). Both FOXP3 and CEACAM6 showed no significant associations with patient age, sex, or tumor location (P>0.05). Notably, elevated expression of both FOXP3 and CEACAM6 was observed in advanced-stage (III-IV) tumors (P<0.001) and poorly differentiated colon cancer (P<0.001) (Fig. 1). These results suggested that FOXP3-positive regulatory T cells and CEACAM6 may contribute to tumor progression and aggressive biological behavior in colon cancer.
Table IIIRelationship between clinicopathological parameters and CEACAM6 and FOXP3 expression levels in patients with colon cancer. |
Significant associations were observed between CEACAM6 expression and T cell infiltration markers (CD3, CD4, CD8, CD45RO and FOXP3) in patients with colon cancer (P<0.05) (Table IV). These findings suggested that CEACAM6 expression is significantly correlated with altered T cell infiltration patterns in colon cancer, particularly showing a strong inverse association with CD3, CD8 and CD45RO but a positive association with FOXP3.
Table IVRelationship between CEACAM6 and CD3, CD4, CD8, CD45RO and FOXP3 expression levels in patients with colon cancer. |
A total of 50 tumor blocks were randomly selected from 301 paraffin-embedded specimens for mRNA extraction, with successful RNA isolation achieved in 35 cases (16 stage I-II and 19 stage III-IV samples). Consistent with IHC findings, transcript levels of CD3, CD8 and CD45RO were significantly downregulated in advanced-stage (III-IV) tumors compared with early-stage (I-II) lesions (P<0.001; Fig. 2). By contrast, CD4 mRNA expression showed no significant intergroup difference (P=0.457; Fig. 3). Both CEACAM6 and FOXP3 mRNA levels were significantly elevated in advanced-stage tumors (III-IV) relative to early-stage disease (P<0.001; Fig. 2).
In the present study, patient follow-up was conducted until June 30, 2025, with OS as the primary endpoint (maximum follow-up duration: 118 months). Kaplan-Meier analysis demonstrated that patients with weakly positive or negative expression of CD3 (median OS: 90 months) and CD45RO (median OS: 85 months) exhibited significantly shorter survival compared with those with strongly positive expression (P=0.001 and P<0.001; Fig. 3). By contrast, strong positivity for CD4 (median OS: 59 months), CEACAM6 (median OS: 35 months), and FOXP3 (median OS: 36.5 months) was associated with poorer outcomes relative to weak/negative expression (P<0.001; Fig. 3).
Kaplan-Meier survival analysis revealed significant prognostic differences based on tumor differentiation and TNM staging. Patients with poorly differentiated tumors demonstrated significantly worse outcomes, with a median OS of 40 months compared with those with well-to-moderately differentiated tumors (P<0.001; Fig. 4). Similarly, advanced-stage (III-IV) cases showed significantly reduced survival (median OS: 49 months) relative to early-stage (I-II) patients (P<0.001; Fig. 4). These findings underscore the strong association between pathological grade, clinical stage and survival outcomes in this cohort.
Univariate Cox regression analysis identified multiple significant prognostic factors for colon cancer outcomes (Table V). The statistically significant variables from univariate Cox analysis were then included in multivariate Cox regression modeling. This analysis identified four independent prognostic factors (Fig. 5): TNM staging [hazard ratio (HR)=4.437; 95% confidence interval (CI): 2.142-9.189; P<0.001), tumor differentiation (HR=2.425; 95% CI: 1.635-3.600; P<0.001), CEACAM6 expression (HR=9.516; 95% CI: 4.133-21.914; P<0.001) and FOXP3 levels (HR=3.345; 95% CI: 1.572-7.118; P=0.002).
The antitumor immune response plays a pivotal role in colon cancer progression, with T cell infiltration serving as a key determinant of clinical outcomes (21). Colon tumors are generally immunogenic and frequently exhibit infiltration by T lymphocytes (22). Multiple studies have shown that reduced T cell infiltration in colon tumors is inversely associated with disease stage and predicts improved OS (23,24). Our findings in poorly differentiated tumors and advanced pathological stages further support this observation. Specifically, it was found that increased infiltration of CD3+, CD8+ and CD45RO+ T cells was associated with improved prognosis, whereas CD4+ T cells showed paradoxical associations. However, the present data highlight limitations of current immune score systems by demonstrating that CEACAM6 and FOXP3 refine prognostic stratification beyond TIL density. For example, patients with tumors exhibiting high CEACAM6 or FOXP3 expression exhibited worse outcomes regardless of CD8+ T-cell levels, suggesting that biomarker panels integrating immune checkpoints and stromal factors may improve risk prediction (16).
FOXP3 is a highly specific marker for Tregs (25). Tumors frequently exhibit increased Treg infiltration, which contributes to immunosuppression by dampening anti-tumor immune responses (26). Multiple studies have demonstrated that elevated CD4+CD25+FOXP3+ Treg infiltration correlates with poor prognosis in various malignancies (27,28). In the present study, transcriptional analysis revealed significant downregulation of CD3, CD8 and CD45RO mRNA levels in advanced-stage tumors, contrasting with the upregulation of CEACAM6 and FOXP3 expression. This differential expression pattern further supports their respective roles in tumor suppression and disease progression. Importantly, multivariate analysis identified CEACAM6 and FOXP3 as independent prognostic factors, underscoring their potential for refining risk stratification in clinical practice. These findings align with recent studies demonstrating that FOXP3+ Treg enrichment in advanced-stage tumors highlights their therapeutic potential as targets for novel immunotherapies designed to disrupt immunosuppressive networks (29,30).
The strong association between CEACAM6 overexpression and advanced tumor stage, poor differentiation, and reduced survival aligns with its documented roles in metastasis and therapy resistance (12). Our findings build upon previous observations by revealing an inverse correlation between CEACAM6 expression and tumor-infiltrating CD3+, CD8+ and CD45RO+ lymphocytes, suggesting its potential role in mediating immune exclusion - a key immunological feature associated with tumor aggressiveness. Mechanistically, CEACAM6 has been shown to contribute to an immunosuppressive microenvironment through multiple pathways. Experimental evidence indicates that CEACAM6 can facilitate the recruitment of MDSCs through interactions with CEACAM1 on immune cells (8). Furthermore, CEACAM6 signaling through Src family kinases and PI3K/AKT pathways can lead to the upregulation of PD-L1 expression on tumor cells (12,15). These mechanisms were demonstrated in murine models where CEACAM6 overexpression resulted in increased PD-L1 levels and MDSC infiltration, while a CEACAM6-targeted vaccine combined with anti-PD-1 antibody synergistically enhanced antitumor immunity (15). These findings position CEACAM6 as a dual biomarker and therapeutic target, particularly for immune checkpoint inhibitor-resistant cases.
The independent prognostic significance of CEACAM6 and FOXP3 supports their integration into clinical decision-making. Patients exhibiting overexpression of either CEACAM6 or FOXP3 may benefit from intensified adjuvant therapy such as anti-CEACAM6 CAR-T cells or antibody-drug conjugates (31,32), or agents modulating Treg function. Targeting CEACAM6 holds promise but requires caution due to its expression on normal granulocytes and epithelial cells, potentially leading to on-target/off-tumor toxicities such as neutropenia or dermatitis (12). Targeting FOXP3+ Tregs systemically carries the risk of inducing autoimmune adverse events. Strategies focusing on specific Treg depletion within the tumor via CTLA-4 inhibition or targeting activation markers might offer improved windows of safety (27,28). Combining CEACAM6 or FOXP3/Treg-targeted strategies with PD-1/PD-L1 blockade represents a rational approach to overcome immune resistance (31). Conversely, tumors lacking both biomarkers but showing high CD8+ T-cell infiltration may be suitable for de-escalation strategies. Future studies should investigate dynamic changes in these biomarkers during treatment to guide adaptive therapeutic approaches.
Our findings on the role of CEACAM6 in promoting immune suppression are mechanistically supported by the study of Pinkert et al (8), which showed that inhibiting the CEACAM6-CEACAM1 interaction potentiates T cell-mediated cytotoxicity. Similarly, Li et al (9) identified CEACAM6 as a critical driver of lung cancer metastasis, underscoring its importance in advanced-stage disease across cancer types. Our observations regarding FOXP3+ Tregs align with clinical evidence from Martinez-Rios et al (26), who demonstrated Treg-associated immunosuppression in colorectal cancer, further supporting the conserved nature of this mechanism. It should be noted, however, that the exclusive use of a patient cohort from Chinese tertiary hospitals represents a limitation of the present study, potentially restricting the generalizability of our conclusions to other ethnic and geographic populations. Variations in genetic background, environmental exposures and regional clinical practices may influence tumor immune biology and biomarker performance. Therefore, large-scale, prospective multinational studies incorporating diverse ethnicities and geographic regions will be essential to validate the universal prognostic value of CEACAM6 and FOXP3 and to establish globally applicable clinical thresholds.
Our multicenter study establishes CEACAM6 and FOXP3 as robust, independent prognostic biomarkers in colon cancer, validated through integrated IHC and transcriptomic analyses. The findings not only corroborate emerging evidence on their roles in tumor progression and immune evasion but also reveal novel mechanistic insights with direct clinical implications. CEACAM6 potentially contributes to an immunosuppressive microenvironment by recruiting MDSCs and upregulating PD-L1(15), providing a mechanistic rationale for the observed immune exclusion and suggesting combination immunotherapy strategies targeting these pathways alongside CEACAM6 itself (32).
While our multicenter design enhances generalizability, limitations include the retrospective nature which limited the availability of consistent data on adjuvant therapies, comorbidities and molecular subtypes such as microsatellite instability status, factors that should be incorporated into future prospective analyses and RNA extraction challenges from FFPE samples (35/50 success rate). The mRNA subset was randomly selected, mitigating selection bias, but future studies should utilize more robust platforms like NanoString or digital spatial profiling for transcriptomic analysis, the latter also enabling spatial resolution of interactions within the tumor microenvironment. Prospective validation using liquid biopsies or fresh-frozen specimens is warranted. Additionally, mechanistic studies, potentially utilizing single-cell RNA sequencing or spatial transcriptomics to precisely map cellular interactions, are needed to clarify whether CEACAM6 directly modulates FOXP3+ Treg recruitment or vice versa.
In conclusion, CEACAM6 and FOXP3 were established as critical biomarkers for colon cancer prognostication, with dual roles in immune evasion and tumor progression. Their incorporation into clinical practice could optimize risk stratification and therapeutic targeting, particularly in the era of precision immunotherapy.
Not applicable.
Funding: The present study was supported by the National Natural Science Foundation of China (grant no. 81172166).
The data generated in the present study may be requested from the corresponding author.
YYL and XXP conceived and designed the study, contributed to data collection and analysis, participated in the interpretation of results, and critically revised the manuscript. YYL prepared the initial draft of the manuscript. JXY contributed to the development of the research methodology and provided supervision throughout the study. JZM carried out a part of the data analysis. YYL, JXY, JZM and XXP confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
The present retrospective study analyzed anonymized data and was approved by the research ethics committees of the Affiliated Hospital of Jiangnan University (approval no. 2025-154; Wuxi, China), the First Affiliated Hospital of Soochow University (approval no. 2025-140; Suzhou, China) and Jiangsu Provincial Veterans Hospital (approval no. 2025-001; Wuxi, China). Written informed consent was obtained from all participants for the use of their tissues in scientific research.
Not applicable.
The authors declare that they have no competing interests.
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Biller LH and Schrag D: Diagnosis and treatment of metastatic colorectal cancer: A review. JAMA. 325:669–685. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Moreno V, Salazar R and Gruber SB: The prognostic value of TILs in stage III colon cancer must consider sidedness. Ann Oncol. 33:1094–1096. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Soeratram TTD, Beentjes I, Egthuijsen JMP, Mookhoek A, Lange MM, Meershoek-Klein Kranenbarg E, Hartgrink HH, van de Velde CJH, Ylstra B, van Laarhoven HWM and van Grieken NCT: A biopsy-based immunoscore in patients with treatment-naive resectable gastric cancer. Ther Adv Med Oncol. 16(17588359241287747)2024.PubMed/NCBI View Article : Google Scholar | |
|
Doi S, Yasuda S, Miyashita M, Nagai M, Nakamura K, Matsuo Y, Terai T, Kohara Y, Sakata T and Sho M: Prognostic relevance of sarcopenia and tumor-infiltrating CD8(+) T cells in patients with hepatocellular carcinoma. Ann Gastroenterol Surg. 9:359–368. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Kong D, Gao C, Yu Y, Yang L, Ma J, Tang S, Mao Y, Li Y and Li N: The distribution characteristics of PD-1 pathway-related immune cells in esophageal cancer tissue and their prognostic significance. PLoS One. 20(e0325349)2025.PubMed/NCBI View Article : Google Scholar | |
|
Zhang C, Chen F, Li J, He Y, Sun J, Zheng Z, Liu G, Wang Y, Kang W and Ye X: Comprehensive analysis of single-cell and bulk RNA sequencing data unveils antigen-presenting and processing fibroblasts and establishes a predictive model in gastric cancer. Cancer Cell Int. 25(225)2025.PubMed/NCBI View Article : Google Scholar | |
|
Pinkert J, Boehm HH, Trautwein M, Doecke WD, Wessel F, Ge Y, Gutierrez EM, Carretero R, Freiberg C, Gritzan U, et al: T cell-mediated elimination of cancer cells by blocking CEACAM6-CEACAM1 interaction. Oncoimmunology. 11(2008110)2022.PubMed/NCBI View Article : Google Scholar | |
|
Li Y, Polyak D, Lamsam L, Connolly ID, Johnson E, Khoeur LK, Andersen S, Granucci M, Stanley G, Liu B, et al: Comprehensive RNA analysis of CSF reveals a role for CEACAM6 in lung cancer leptomeningeal metastases. NPJ Precis Oncol. 5(90)2021.PubMed/NCBI View Article : Google Scholar | |
|
Huskey ALW, McNeely I and Merner ND: CEACAM gene family mutations associated with inherited breast cancer risk-A comparative oncology approach to discovery. Front Genet. 12(702889)2021.PubMed/NCBI View Article : Google Scholar | |
|
Ilantzis C, DeMarte L, Screaton RA and Stanners CP: Deregulated expression of the human tumor marker CEA and CEA family member CEACAM6 disrupts tissue architecture and blocks colonocyte differentiation. Neoplasia. 4:151–163. 2002.PubMed/NCBI View Article : Google Scholar | |
|
Wu G, Wang D, Xiong F, Wang Q, Liu W, Chen J and Chen Y: The emerging roles of CEACAM6 in human cancer (Review). Int J Oncol. 64(27)2024.PubMed/NCBI View Article : Google Scholar | |
|
Wang X, Yang C, Wang X and Duan P: Pan-cancer analysis reveals a regulatory pattern of anoikis in human cancers. Cell Mol Biol (Noisy-le-grand). 70:51–61. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Zang M, Zhang B, Zhang Y, Li J, Su L, Zhu Z, Gu Q, Liu B and Yan M: CEACAM6 promotes gastric cancer invasion and metastasis by inducing epithelial-mesenchymal transition via PI3K/AKT signaling pathway. PLoS One. 9(e112908)2014.PubMed/NCBI View Article : Google Scholar | |
|
Li Y, Zhu X, You J, Zhang B, Huang X and Jin C: Efficacy of bivalent CEACAM6/4-1BBL genetic vaccine combined with anti-PD1 antibody in MC38 tumor model of mice. Heliyon. 8(e10775)2022.PubMed/NCBI View Article : Google Scholar | |
|
Liu Y, Xia T, Jin C, Gu D, Yu J, Shi W, Zhang KE, Zhang L, Ye J and Li L: FOXP3 and CEACAM6 expression and T cell infiltration in the occurrence and development of colon cancer. Oncol Lett. 11:3693–3701. 2016.PubMed/NCBI View Article : Google Scholar | |
|
Karamchandani DM, Gonzalez RS, Lee H, Westerhoff M, Cox B and Pai RK: Interobserver agreement and practice patterns for grading of colorectal carcinoma: World health organization (WHO) classification of tumours 5th edition vs. American joint committee on cancer (AJCC) 8th edition staging manual. Histopathology. 86:1101–1111. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F and Cree IA: The 2019 WHO classification of tumours of the digestive system. Histopathology. 76:182–188. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Condurache Hritcu OM, Ciobanu Apostol DG, Toader SV, Solcan C, Brănișteanu DE, Toader MP and Costan VV: Immunohistochemical assessment of maspin, β-catenin, and MMP-14 in oral potentially malignant lesions and oral squamous cell carcinoma: A retrospective observational study. Medicina (Kaunas). 61(1037)2025.PubMed/NCBI View Article : Google Scholar | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Menard LC, Fischer P, Kakrecha B, Linsley PS, Wambre E, Liu MC, Rust BJ, Lee D, Penhallow B, Manjarrez Orduno N and Nadler SG: Renal cell carcinoma (RCC) tumors display large expansion of double positive (DP) CD4+CD8+ T cells with expression of exhaustion markers. Front Immunol. 9(2728)2018.PubMed/NCBI View Article : Google Scholar | |
|
Huang CY, Chiang SF, Ke TW, Chen TW, You YS, Chen WT and Chao KSC: Clinical significance of programmed death 1 ligand-1 (CD274/PD-L1) and intra-tumoral CD8+ T-cell infiltration in stage II-III colorectal cancer. Sci Rep. 8(15658)2018.PubMed/NCBI View Article : Google Scholar | |
|
Thomas CE, Takashima Y, Buchanan DD, Wesselink E, Qu C, Hsu L, Dias Costa A, Gallinger S, Grant RC, Huyghe JR, et al: Density of T-cell subsets in colorectal cancer in relation to disease-specific survival. Cancer Epidemiol Biomarkers Prev. 34:1122–1133. 2025.PubMed/NCBI View Article : Google Scholar | |
|
Küçükköse E, Baars MJD, Amini M, Schraa SJ, Floor E, Bol GM, Borel Rinkes IHM, Roodhart JML, Koopman M, Laoukili J, et al: Stromal localization of inactive CD8(+) T cells in metastatic mismatch repair deficient colorectal cancer. Br J Cancer. 130:213–223. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Prasongtanakij S, Soontrapa K and Thumkeo D: The role of prostanoids in regulatory T cells and their implications in inflammatory diseases and cancers. Eur J Cell Biol. 104(151482)2025.PubMed/NCBI View Article : Google Scholar | |
|
Martinez-Rios J, Lopez-Pacheco CP, Garcia-Zepeda EA and Soldevila G: CCR9 shapes the immune microenvironment of colorectal cancer modulating the balance between intratumoral CD8+ T cell and FoxP3+ Helios+ Treg subpopulations. PLoS One. 20(e0321930)2025.PubMed/NCBI View Article : Google Scholar | |
|
Tanaka A and Sakaguchi S: Targeting treg cells in cancer immunotherapy. Eur J Immunol. 49:1140–1146. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Tanaka A and Sakaguchi S: Regulatory T cells in cancer immunotherapy. Cell Res. 27:109–118. 2017.PubMed/NCBI View Article : Google Scholar | |
|
Shah F, Giri PS, Bharti AH and Dwivedi M: Compromised melanocyte survival due to decreased suppression of CD4(+) & CD8(+) resident memory T cells by impaired TRM-regulatory T cells in generalized vitiligo patients. Exp Dermatol. 33(e14982)2024.PubMed/NCBI View Article : Google Scholar | |
|
Panek WK, Toedebusch RG, McLaughlin BE, Dickinson PJ, Van Dyke JE, Woolard KD, Berens ME, Lesniak MS, Sturges BK, Vernau KM, et al: The CCL2-CCR4 axis promotes regulatory T cell trafficking to canine glioma tissues. J Neurooncol. 169:647–658. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Redin E, Garmendia I, Lozano T, Serrano D, Senent Y, Redrado M, Villalba M, De Andrea CE, Exposito F, Ajona D, et al: SRC family kinase (SFK) inhibitor dasatinib improves the antitumor activity of anti-PD-1 in NSCLC models by inhibiting treg cell conversion and proliferation. J Immunother Cancer. 9(e001496)2021.PubMed/NCBI View Article : Google Scholar | |
|
Nakazawa Y, Miyano M, Tsukamoto S, Kogai H, Yamamoto A, Iso K, Inoue S, Yamane Y, Yabe Y, Umihara H, et al: Delivery of a BET protein degrader via a CEACAM6-targeted antibody-drug conjugate inhibits tumour growth in pancreatic cancer models. Nat Commun. 15(2192)2024.PubMed/NCBI View Article : Google Scholar |