bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–04–20
seventeen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Int J Gynecol Cancer. 2024 Dec;pii: S1048-891X(24)22175-7. [Epub ahead of print]34(12): 1853-1860
       OBJECTIVE: To assess the relation between immune microenvironment, survival, and clinicopathological characteristics.
    METHODS: This study was a retrospective, single-center, observational study. Patients with a vulvovaginal melanoma and available archived material were included. All cases underwent pathology review, tumor-infiltrating lymphocyte quantification, and next-generation sequencing analysis, when feasible. Clinical data included demographic, treatment, and prognostic data.
    RESULTS: Forty-two patients were selected during the study period, but 13 were finally excluded owing to unavailable formalin-fixed, paraffin-embedded material or unknown follow-up data. Twelve of 19 cases (63.2%) had at least one genetic mutation, 3/18 (16.7%) had BRAF, 3/18 (16.7%) had c-KIT mutation, and 4/17 (23.5%) had NRAS mutations. High stromal tumor-infiltrating lymphocytes were identified in 13/28 patients (46.4%), and brisk tumor-infiltrating lymphocytes in 17/28 patients (60.7%). A density of stromal tumor-infiltrating lymphocytes >40% and brisk distribution were the single clinicopathologic factor associated with increased disease-free survival.
    CONCLUSION: The study showed that brisk tumor-infiltrating lymphocytes and stromal tumor-infiltrating lymphocytes were a marker for disease progression, and for response to immunotherapy strategies. To validate these findings on a larger scale, further research is warranted through a multicenter study with a larger cohort and additional genetic and translational analysis.
    Keywords:  Melanoma; Pathology; Vulvar Neoplasms; Vulvar and Vaginal Cancer
    DOI:  https://doi.org/10.1136/ijgc-2024-005359
  2. J Adv Pract Oncol. 2025 Mar 16. 1-14
      Adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) was recently approved for patients with advanced melanoma (metastatic or unresectable) previously treated with immune checkpoint inhibitors and BRAF/MEK targeted therapies (where appropriate). Tumor-infiltrating lymphocytes isolated from patient-derived tumor tissues enter the tumor microenvironment and recognize tumor-specific antigens, leading to the destruction of tumor cells. The multistep TIL cell therapy journey is led by a multidisciplinary health care team. Patients selected for TIL cell therapy undergo tumor tissue procurement for TIL generation, followed by preparative lymphodepletion before receiving a single-dose infusion of TIL and a short course of high-dose interleukin-2. Successful implementation of TIL cell therapy requires well-established procedures and workflows to select and screen patients, procure tumor tissue, administer TIL cell therapy, and monitor patients during treatment and after discharge. The advanced practice provider plays a central role in a patient's TIL treatment journey by planning and coordinating care across the health-care system, educating patients and staff, and providing direct and supportive patient care. Here, we review the treatment landscape for advanced melanoma and clinical data supporting TIL cell therapy. We also provide guidance related to patient selection, tumor tissue procurement, TIL cell therapy regimen, safety monitoring, symptom management, and post-discharge follow-up.
    DOI:  https://doi.org/10.6004/jadpro.2025.16.7.8
  3. Front Immunol. 2025 ;16 1581098
      Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality globally. The tumor microenvironment (TME) plays a pivotal role in HCC progression, characterized by dynamic interactions between stromal components, immune cells, and tumor cells. Key immune players, including tumor-associated macrophages (TAMs), tumor-infiltrating lymphocytes (TILs), cytotoxic T lymphocytes (CTLs), regulatory T cells (Tregs), MDSCs, dendritic cells (DCs), and natural killer (NK) cells, contribute to immune evasion and tumor progression. Recent advances in immunotherapy, such as immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell therapy (ACT), and combination therapies, have shown promise in enhancing anti-tumor responses. Dual ICI combinations, ICIs with molecular targeted drugs, and integration with local treatments or radiotherapy have demonstrated improved outcomes in HCC patients. This review highlights the evolving understanding of the immune microenvironment and the therapeutic potential of immunotherapeutic strategies in HCC management.
    Keywords:  CAR-T therapy; HCC; NK cell therapy; immune microenvironment; immunotherapy
    DOI:  https://doi.org/10.3389/fimmu.2025.1581098
  4. J Pathol Inform. 2025 Apr;17 100437
      The prognostic relevance of tumor-infiltrating lymphocytes (TILs) in non-small cell Lung cancer (NSCLC) is well-established. However, manual TIL quantification in hematoxylin and eosin (H&E) whole slide images (WSIs) is laborious and prone to variability. To address this, we aim to develop and validate an automated computational pipeline for the quantification of TILs in WSIs of NSCLC. Such a solution in computational pathology can accelerate TIL evaluation, thereby standardizing the prognostication process and facilitating personalized treatment strategies. We develop an end-to-end automated pipeline for TIL estimation in Lung cancer WSIs by integrating a patch extraction approach based on hematoxylin component filtering with a machine learning-based patch classification and cell quantification method using the HoVer-Net model architecture. Additionally, we employ randomized patch sampling to further reduce the processed patch amount. We evaluate the effectiveness of the patch sampling procedure, the pipeline's ability to identify informative patches and computational efficiency, and the clinical value of produced scores using patient survival data. Our pipeline demonstrates the ability to selectively process informative patches, achieving a balance between computational efficiency and prognostic integrity. The pipeline filtering excludes approximately 70% of all patch candidates. Further, only 5% of eligible patches are necessary to retain the pipeline's prognostic accuracy (c-index = 0.65), resulting in a linear reduction of the total computational time compared to the filtered patch subset analysis. The pipeline's TILs score has a strong association with patient survival and outperforms traditional CD8 immunohistochemical scoring (c-index = 0.59). Kaplan-Meier analysis further substantiates the TILs score's prognostic value. This study introduces an automated pipeline for TIL evaluation in Lung cancer WSIs, providing a prognostic tool with potential to improve personalized treatment in NSCLC. The pipeline's computational advances, particularly in reducing processing time, and clinical relevance demonstrate a step forward in computational pathology.
    Keywords:  Automated quantification; Computational pathology; Deep learning; Explainable AI; Frugal AI; Machine learning; Non-small cell Lung cancer; Resource-efficient AI; Tumor-infiltrating lymphocytes; Whole slide images
    DOI:  https://doi.org/10.1016/j.jpi.2025.100437
  5. Int J Gynecol Cancer. 2024 Apr;pii: S1048-891X(24)01343-4. [Epub ahead of print]34(4): 490-496
       BACKGROUND: Data suggest an association between positron emission tomography/CT (PET/CT) metabolic metrics and tumor microenvironment in several malignancies, and a potential role of PET/CT to monitor response to immunotherapy.
    OBJECTIVE: To evaluate the correlation between tumor loco-regional extension and tumor-infiltrating lymphocyte infiltration in locally advanced cervical cancer prior to concurrent chemo-radiotherapy. The secondary objective was to assess the association between tumor-infiltrating lymphocytes and PET/CT metabolic metrics.
    METHODS: Patients with locally advanced cervical cancer and negative para-aortic extensions on PET/CT were included. Two senior nuclear medicine physicians specializing in gynecologic oncology reviewed all PET/CT exams, and extracted tumor maximum standardized uptake value, metabolic tumor volume, and total lesion glycolysis, as well as pelvic lymph node involvement. One senior gynecologic oncology pathologist assessed intraepithelial tumor-infiltrating lymphocytes and stromal tumor-infiltrating lymphocytes. Intraepithelial tumor-infiltrating lymphocytes were categorized following previous studies as <1% and >1%. The cut-off for stromal tumor-infiltrating lymphocytes was chosen empirically: intermediate <60% and high >60%.
    RESULTS: 86 patients were included. Intraepithelial tumor-infiltrating lymphocytes were not significantly associated with tumor metabolic metrics. Intraepithelial tumor-infiltrating lymphocytes were not significantly associated with maximum standard uptake value (p=0.16), or metabolic tumor volume (p=0.19). Tumors with <1% intraepithelial tumor-infiltrating lymphocytes score were associated with a higher MRI tumor size (≥ median) (63.3% vs 39.3%, p=0.04). Patients with pelvic lymph node uptake were significantly more frequent in patients with high stromal tumor-infiltrating lymphocytes score (≥60%) (61.5% vs 31.7%, p=0.009).
    CONCLUSIONS: Poor or absent intraepithelial tumor-infiltrating lymphocytes were associated with more advanced disease at diagnosis and larger tumor size. Tumor-infiltrating lymphocytes were not associated with tumor metabolic activity. Intraepithelial and stroma tumor-infiltrating lymphocytes are not redundant and should be assessed separately. Further work is needed to evaluate the association between tumor metabolic profile and immune populations, including different T-cell subtypes for patient selection for immunotherapy strategies.
    Keywords:  cervical cancer; lymph nodes
    DOI:  https://doi.org/10.1136/ijgc-2023-004677
  6. Am J Cancer Res. 2025 ;15(3): 1335-1345
      The immune system's role in cancer development and progression is receiving increasing attention. Endometrial cancer, common gynecological malignancy, has exhibited promising responses to immunotherapies. This study aims to assess the prognostic significance of various immune cell subsets in endometrial cancer, focusing on potential novel biomarkers and therapeutic targets. A systematic literature review and meta-analysis were conducted. Eleven eligible studies, comprising 2,319 patients with endometrial cancer, were included. The primary outcome was the association between levels of immune cell types, particularly CD8+ T cells, and overall prognosis. The meta-analysis found that high levels of tumor-infiltrating lymphocytes (TILs), particularly CD8+ T cells, were significantly associated with better overall prognosis in endometrial cancer patients. These findings suggest that the tumor immune microenvironment plays a crucial role in endometrial cancer prognosis. This meta-analysis indicates that higher levels of CD8+ T cells in the tumor microenvironment are linked to improved prognosis in endometrial cancer, underscoring the immune system's potential in prognostication and therapy.
    Keywords:  Immune cell infiltration; endometrial cancer; personalized treatment; prognostic biomarkers; tumor microenvironment
    DOI:  https://doi.org/10.62347/BXZM8857
  7. Braz J Med Biol Res. 2025 ;pii: S0100-879X2025000100633. [Epub ahead of print]58 e14467
      Immune checkpoint blockade with anti-programmed cell death protein 1 (PD-1) antibody has become a hot topic for the treatment of human malignancies. Here, we aimed to investigate whether the percentage of PD-1 in CD8+ tumor-infiltrating lymphocytes correlates with the progression of colonic-derived peritoneal adenocarcinoma (PA). Peripheral blood and tissue samples from 40 patients with colonic-derived PA were collected and subjected to multicolor flow cytometry analysis of the percentage of peripheral PD-1+CD8+ T cells. The multiple immunofluorescence method was used to detect the positive percentages of PD-1 and CD8 in the tissues. The enrolled patients were divided into groups by recurrence interval (less than 6 months, greater than two years) and differentiation grade (low, well/moderate). In the colonic-derived PA tissues, the percentages of cells positive for PD-1, CD8, and PD-1+CD8+ were higher in the paracancer tissues compared with cancerous tissues. PD-1+CD8+ T cells had an increased presence in peripheral blood than in tissues. Our data also indicated that colonic-derived PA patients with less than a six-month recurrence interval presented higher levels of PD-1 in CD8+ tumor-infiltrating lymphocytes in than the two-year recurrence group. The level of PD-1+CD8+T cells in the tissue correlated with the clinical outcome of colonic-derived PA. Higher percentages of PD-1+CD8+T cells correlated with a shorter progression-free survival (PFS). PD-1 in CD8+ tumor-infiltrating lymphocytes may have a good predictive value for immunotherapy of colonic-derived PA and act as the prognostic factor for PFS.
    DOI:  https://doi.org/10.1590/1414-431X2025e14467
  8. Cancer Med. 2025 Apr;14(8): e70445
       AIM: With advances in systemic therapy, the number of patients with hepatocellular carcinoma (HCC) who can undergo hepatic resection has increased in recent years, but there are no reports evaluating the immune status in the peritumoral area.
    METHODS: We enrolled 14 patients who underwent hepatic resection after lenvatinib (LEN, n = 7) or atezolizumab plus bevacizumab (ATZ/BEV, n = 5) therapy. Tumor-infiltrating lymphocytes (TILs), including CD3+ and CD8+ TILs, in the peritumoral area were evaluated by hematoxylin and eosin staining and immunohistochemistry.
    RESULTS: The median TIL counts after LEN and ATZ/BEV therapy were 32 and 92 cells/0.237 mm2, respectively (p = 0.0044). The median CD3+ TIL counts after LEN and ATZ/BEV therapy were 26 and 71 cells/0.237 mm2, respectively (p = 0.0057). The median CD8+ TIL counts after LEN and ATZ/BEV therapy were 14 and 42 cells/0.237 mm2, respectively (p = 0.0044).
    CONCLUSION: TIL counts, including those of CD3+ and CD8+ TILs, in the peritumoral area were significantly higher after ATZ/BEV than after LEN therapy.
    Keywords:  atezolizumab plus bevacizumab; conversion surgery; hepatocellular carcinoma; lenvatinib
    DOI:  https://doi.org/10.1002/cam4.70445
  9. J Immunother Cancer. 2025 Apr 17. pii: e010736. [Epub ahead of print]13(4):
       BACKGROUND: SOT201 and its murine surrogate mSOT201 are novel cis-acting immunocytokines consisting of a humanized/murinized/, Fc-silenced anti-programmed cell death protein 1 (PD-1) monoclonal antibody (mAb) fused to an attenuated human interleukin (IL)-15 and the IL-15Rα sushi+ domain. Murine mPD1-IL2v is a conjugate of a murinized, Fc silenced anti-PD-1 mAb bearing human IL-2 with abolished IL-2Rα binding. These immunocytokines spatiotemporally reinvigorate PD-1+ CD8+ tumor-infiltrating lymphocytes (TILs) via cis-activation and concomitantly activate the innate immunity via IL-2/15Rβγ signaling.
    METHODS: Human peripheral blood mononuclear cell and cell lines were used to evaluate cis/trans activity of SOT201. Anti-PD-1 mAb responsive (MC38, CT26) and resistant (B16F10, CT26 STK11 KO) mouse tumor models were used to determine the anticancer efficacy, and the underlying immune cell activity was analyzed via single-cell RNA sequencing and flow cytometry. The expansion of tumor antigen-specific CD8+ T cells by mSOT201 or mPD1-IL2v and memory CD8+ T-cell generation in vivo was determined by flow cytometry.
    RESULTS: SOT201 delivers attenuated IL-15 to PD-1+ T cells via cis-presentation, reinvigorates exhausted human T cells and induces higher interferon-γ production than pembrolizumab in vitro. mSOT201 administered as a single dose exhibits strong antitumor efficacy with several complete responses in all tested mouse tumor models. While mPD1-IL2v activates CD8+ T cells with a 50-fold higher potency than mSOT201 in vitro, mSOT201 more effectively reactivates effector exhausted CD8+ T cells (Tex), which demonstrate higher cytotoxicity, lower exhaustion and lower immune checkpoint transcriptional signatures in comparison to mPD1-IL2v in MC38 tumors in vivo. This can be correlated with a higher rate of complete responses in the MC38 tumor model following mSOT201 treatment when compared with mPD1-IL2v. mSOT201 increased the relative number of tumor antigen-specific CD8+ T cells, and unlike mPD1-IL2v stimulated greater expansion of adoptively transferred ovalbumin-primed CD8+ T cells simultaneously limiting the peripheral CD8+ T-cell sink, leading to the development of memory CD8+ T cells in vivo.
    CONCLUSIONS: SOT201 represents a promising therapeutic candidate that preferentially targets PD-1+ TILs, delivering balanced cytokine activity for reviving CD8+ Tex cells in tumors. SOT201 is currently being evaluated in the Phase I clinical study VICTORIA-01 (NCT06163391) in patients with advanced metastatic cancer.
    Keywords:  Cytokine; Immune Checkpoint Inhibitor; Immunotherapy; T cell; Tumor microenvironment - TME
    DOI:  https://doi.org/10.1136/jitc-2024-010736
  10. Am J Cancer Res. 2025 ;15(3): 976-990
      Head and neck carcinomas are the sixth most common cancers worldwide, with laryngeal squamous cell carcinoma (LSCC) being the second most prevalent subtype. Improving survival outcomes in LSCC patients remains a critical clinical challenge. This retrospective study aimed to develop a nomogram model integrating tumor-infiltrating lymphocytes (TILs) and clinicopathological characteristics to predict the prognosis of LSCC patients. The nomogram model was constructed using Cox and Lasso regression analyses and was subsequently evaluated through receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA). Data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were utilized for model validation and to further elucidate the role of TILs and immune responses in LSCC. This study cohort included LSCC patients diagnosed by pathological examination between 2011 and 2014 at Xiangya Hospital and Harbin Medical University Cancer Hospital. A total of 412 patients were assigned to the training cohort and 140 patients to the test cohort for validation. The final nomogram model integrated TNM stage, TILs, PLR, BMI, age, differentiation and NLR. The area under the curve (AUC) was 0.745, indicating strong calibration and clinical utility. Kaplan-Meier survival curves demonstrated significant discrimination. TILs were positively correlated with immune cell abundance and the expression of immune-related genes. In conclusion, the nomogram model based on TILs and clinicopathological features effectively predicts the prognosis of LSCC patients.
    Keywords:  Prognosis; TNM staging system; laryngeal squamous cell carcinoma; nomogram model; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.62347/MKFI3976
  11. J Transl Med. 2025 Apr 17. 23(1): 453
       BACKGROUND: Patients with small-cell lung cancer (SCLC) have the poor prognosis. Current research suggested that low-dose radiotherapy (LDRT) combined with immunotherapy can enhance the immunogenicity of tumor cells, thereby improving antigen presentation and promoting the intratumoral infiltration of CD8+ T cells, which significantly extends the survival of patients. However, the change trajectory of T cells, and the mechanisms underlying the promotion of intratumoral infiltration of CD8+ T cells, and the enhancement of their cytotoxic functions remain to be elucidated.
    METHODS: To delineate the dynamic changes of T cells, we collected tumors from Kaede tumor-bearing mice that had undergone radioimmunotherapy. Using flow cytometry, we sorted intratumoral-infiltrating immune cells, which were required for single-cell RNA sequencing, at various time points (Kaede Red: derived from tumor-draining lymph node [TDLN]). The results obtained from the sequencing analysis were further validated through experiments, such as flow cytometry, immunofluorescence, and analysis of clinical cohort data.
    RESULTS: Here, we observed stem-like T cells migrating from the TDLN to the tumor site and differentiating into effector phenotypes within the tumor. Dendritic cells (DCs) are the key cluster that induces the differentiation of stem-like T cell into effector phenotypes. Moreover, SCLC patients with a high infiltration of tumor-specific CXCR6+CD8+ T cells exhibited a supportive TME and longer survival time (P < 0.001).
    CONCLUSIONS: This study delineates the change trajectory of CD8+ T cells, identifies the crucial role of DCs in T cell differentiation, and highlights the significance of tumor-specific CXCR6+CD8+ T cells in anti-tumor immunity. Future therapeutic strategies for SCLC could focus on enhancing the infiltration of activated DCs and CXCR6+CD8+ T cells within the tumor microenvironment to improve treatment efficacy.
    Keywords:  CD8-positive T-lymphocytes; Immunotherapy; Radiotherapy; Small-cell lung cancer; Tumor-infiltrating
    DOI:  https://doi.org/10.1186/s12967-025-06450-1
  12. Res Sq. 2025 Apr 01. pii: rs.3.rs-6173056. [Epub ahead of print]
      We developed a deep learning Pathomics image analysis workflow to generate spatial Tumor-TIL maps to visualize and quantify the abundance and spatial distribution of tumor infiltrating lymphocytes (TILs) in colon cancer. Colon cancer and lymphocyte detection in hematoxylin and eosin (H&E) stained whole slide images (WSIs) has revealed complex immuno-oncologic interactions that form TIL-rich and TIL-poor tumor habitats, which are unique in each patient sample. We compute Tumor%, total lymphocyte%, and TILs% as the proportion of the colon cancer microenvironment occupied by intratumoral lymphocytes for each WSI. Kaplan-Meier survival analyses and multivariate Cox regression were utilized to evaluate the prognostic significance of TILs% as a Pathomics biomarker. High TILs% was associated with improved overall survival (OS) and progression-free interval (PFI) in localized and metastatic colon cancer and other clinicopathologic variables, supporting the routine use of Pathomics Tumor-TIL mapping in biomedical research, clinical trials, laboratory medicine, and precision oncology.
    DOI:  https://doi.org/10.21203/rs.3.rs-6173056/v1
  13. Methods Mol Biol. 2025 ;2904 259-271
      Metabolic reprogramming is increasingly recognized as a fundamental aspect of T cell activation, influencing the differentiation, proliferation, and effector functions of lymphocytes. Measuring and screening the metabolic states of activated T cells provide insights into the dynamic interplay between cellular metabolism and immune function. In the following chapter, we provide a simple protocol based on the publication of Argüello et al. [1] to analyze the metabolic state of activated T cells at the single-cell level using standard flow cytometry.
    Keywords:  FACS; Glycolysis; Metabolism; Oxidative phosphorylation; SCENITH; T cells
    DOI:  https://doi.org/10.1007/978-1-0716-4414-0_18
  14. J Immunother Cancer. 2025 Apr 17. pii: e010962. [Epub ahead of print]13(4):
       BACKGROUND: T cells modified to express a chimeric antigen receptor (CAR) are successful against B-lineage malignancies but fail to induce durable remissions in up to half of patients and have shown limited efficacy against other types of cancer. Strategies to improve CAR T cell potency and responses to low antigen densities without inducing CAR T cell dysfunction or limiting persistence are necessary to expand durability of remissions.
    METHODS: We overexpressed T-bet in human and mouse CAR T cells to mimic exposure to signal 3 cytokines during T cell priming to promote T helper cell 1 (Th1) polarization of CD4+CAR T cells with the goal of enhancing antitumor activity. Using human CAR T cells and xenograft models we interrogated the impact of T-bet overexpression on CAR T cell antitumor activity in vitro and in vivo. We also used a syngeneic murine CAR T cell model to study the impact of T-bet overexpression on long-term persistence and secondary responses to tumor rechallenge.
    RESULTS: T-bet overexpression reduced expression of the Th2 cytokine interleukin 4 and promoted polyfunctional production of Th1-associated cytokines in response to CAR stimulation. T-bet overexpression enhanced some effector functions in vitro but did not improve CAR T cell-mediated control of leukemia expressing high levels of antigen in vivo. T-bet overexpression also improved effector function of murine CD19 CAR T cells with no impairment to the persistence or ability of persistent CAR T cells to re-expand and clear a secondary leukemia challenge. Finally, T-bet overexpression promoted enhanced in vitro function against leukemia expressing low levels of CD19, which translated to improved control of CD19lo leukemia in vivo by human C19 CAR T cells containing a 4-1BB costimulatory domain.
    CONCLUSIONS: Together, our data demonstrate that T-bet overexpression induces a reduction in Th2 cytokine production, an increase in polyfunctional Th1 cytokine production and enhances 4-1BB CAR T cell activity against cancers expressing low levels of target antigen without promoting a loss in functional CAR T cell persistence.
    Keywords:  Adoptive cell therapy - ACT; Chimeric antigen receptor - CAR
    DOI:  https://doi.org/10.1136/jitc-2024-010962
  15. Front Immunol. 2025 ;16 1496640
       Background: Colorectal adenocarcinoma (COAD) is a prevalent malignant tumor associated with a high mortality rate. Within the tumor microenvironment, CD8+ T cells play a pivotal role in the anti-tumor immune response within the human body. Fibrosis directly and indirectly affects the therapeutic response of tumor immunotherapy. However, the significance of regulatory genes associated with tumor-associated fibrosis and CD8+ T cell infiltration remains uncertain. Therefore, it is imperative to identify biomarkers with prognostic value and elucidate the precise role of CD8+ T cells and tumor-associated fibrosis.
    Methods: We performed a single-cell transcriptome analysis of COAD samples from the GEO database. To evaluate immune infiltration in COAD samples, we utilized CIBERSORT and ESTIMATE. Furthermore, we analyzed the correlation between CD8+ T cells and immune infiltration. To analyze COAD expression's quantitative immune cell composition data, we conducted a Weighted Gene Correlation Network Analysis and utilized a deconvolution algorithm. The data for these analyses were obtained from the GEO database. We utilized univariate Cox regression and LASSO analysis to create a prognostic model. The predictive model was assessed through Kaplan-Meier analysis, and a survival prediction nomogram was created. Additionally, we analyzed the correlation between the prognostic model and chemotherapy drug sensitivity. To estimate the expression of hub genes, we employed immunohistochemistry, real-time PCR, and western blot techniques.
    Results: Single-cell transcriptome analysis has indicated a higher prevalence of CD8+ T cells in COAD tumor samples. The connection between COAD and CD8+ T cells was further confirmed by WGCNA and deconvolution analysis using the GEO database. The Protein-Protein Interaction network analysis revealed three hub genes: LARS2, SEZ6L2, and SOX7. A predictive model was subsequently created using LASSO and univariate COX regression, which included these three genes. Two of these hub genes (LARS2 and SEZ6L2) were found to be upregulated in COAD cell lines and tissues, while SOX7 was observed to be downregulated. The prognostic model demonstrated a significant association with CD8+ T cells, suggesting that these genes could serve as potential biomarkers and targets for gene therapy in treating COAD.
    Conclusion: This study has identified three key genes associated with CD8+ T cells and the prognosis of COAD, providing new prognostic biomarkers for diagnosing and treating COAD.
    Keywords:  CD8 + T cells; colon adenocarcinoma; fibrosis; prognostic biomarkers; single-cell RNA sequencing
    DOI:  https://doi.org/10.3389/fimmu.2025.1496640