bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–12–28
fifteen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. bioRxiv. 2025 Oct 06. pii: 2025.10.05.680562. [Epub ahead of print]
      T cell exhaustion limits the efficacy of cancer immunotherapies. Here, we performed genome-wide loss-of-function screening in repetitively stimulated human T cells and identified the mulitfunctional ubiquitin-modifying protein A20/TNFAIP3 as a major negative regulator of exhausted T cell persistence. Protein large language modeling, deep base-editing mutagenesis, and studies in immunocompetent mice with domain-specific inactivating mutations revealed A20's non-enzymatic M1 ubiquitin-binding zinc finger 7 (A20 ZF7 ) motif as critical to suppression of anti-tumor immunity. A20 ZF7 -deficient CD8 + tumor-infiltrating lymphocytes (TILs) resisted terminal exhaustion and circumvented an unappreciated mechanism restraining perforin degranulation in terminally exhausted cells. Human chimeric antigen receptor (CAR)-T cells engineered via base-editing to inactivate A20 ZF7 via a single missense mutation also resisted exhaustion, secreted more perforin and robustly suppressed cancer in vivo . These studies pinpoint A20 ZF7 as a novel T cell checkpoint and reveal precision base-editing of missense mutations as an effective approach to enhance CAR-T cell therapy.
    DOI:  https://doi.org/10.1101/2025.10.05.680562
  2. Ther Adv Med Oncol. 2025 ;17 17588359251403410
       Background: The clinical validity of tumor-infiltrating lymphocytes (TILs) has been investigated extensively in triple-negative breast cancer (TNBC). However, inconsistent cutoff values across studies limit its clinical application.
    Objective: To determine the optimal TILs cutoff predicting pathological complete response (pCR) and event-free survival (EFS) in Vietnamese TNBC patients undergoing neoadjuvant chemotherapy (NACT).
    Design: A single-center, retrospective cohort study of 106 stage II-III TNBC patients treated with NACT at Ho Chi Minh City Oncology Hospital between January 2022 and May 2023.
    Methods: Stromal TILs (sTILs) were evaluated on pre-treatment biopsy specimens using the International TILs Working Group guideline. The optimal cutoff was determined using receiver operating characteristic curve analysis. Logistic regression and Cox proportional hazards models were used to analyze potentially predictive and prognostic factors.
    Results: An optimal 20% TILs cutoff stratified the cohort into high- and low-TIL groups, with significantly different pCR rates (50.9% vs 9.8%, p < 0.001). High TILs (odds ratio (OR) = 8.22, p < 0.001) and lower clinical tumor status (OR = 3.64, p = 0.033) independently predicted pCR. TILs (hazard ratio (HR) = 0.38, p = 0.017) and pCR status (HR = 0.08, p = 0.017) were independent prognostic factors for EFS.
    Conclusion: A sTIL cutoff of 20% effectively predicts chemotherapy response and survival outcome in Vietnamese TNBC patients, supporting its use in clinical decision-making.
    Keywords:  event-free survival; optimal TILs cutoff; pathological complete response; predictive factor; prognostic factor; triple-negative breast cancer; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1177/17588359251403410
  3. Cytokine Growth Factor Rev. 2025 Dec 13. pii: S1359-6101(25)00165-0. [Epub ahead of print]
      T cells have traditionally been characterized as autonomous killers that eliminate malignant cells through single-cell cytotoxicity. However, recent findings reveal that tumor-reactive T cells frequently form tightly bound clusters on the tumor surface, functioning as cooperative hyper-effector units. These clusters establish stable and reinforced immune synapses, sustain prolonged engagement with tumor targets, and coordinate the delivery of perforin, granzymes, and proinflammatory cytokines. As a result, they exhibit markedly enhanced tumor-killing capacity compared with unbound T cells in both ex vivo assays and murine models. This discovery introduces an essential spatial and mechanical dimension to cancer immunology and challenges the prevailing assumption that molecular phenotype alone defines T-cell efficacy. The implications are substantial, extending to tumor-infiltrating lymphocyte (TIL) therapy optimization, the refinement of immunotherapy biomarkers, and the engineering of next-generation CAR-T and CAR-NK platforms. Enriching or mimicking the behavior of cluster-forming T cells may accelerate clinical responses and overcome barriers associated with solid tumors. Collectively, these insights reposition T-cell clustering as a fundamental determinant of effective antitumor immunity.
    Keywords:  CAR-T therapy; Digital pathology; Immune synapse; Spatial cancer immunology; T-cell clusters; Tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1016/j.cytogfr.2025.12.007
  4. Cell Rep. 2025 Dec 19. pii: S2211-1247(25)01529-3. [Epub ahead of print]45(1): 116757
      Interleukin-12 (IL-12) is a proinflammatory T cell- and natural killer (NK) cell-activating cytokine with potent preclinical anti-tumor efficacy, but clinical toxicity has limited its use as an immunotherapy. In a mouse tumor model, wild-type IL-12 induces NK cell hyperactivation and cytokine storm followed by rapid NK cell loss. To avoid NK hyperactivation, we engineered an IL-12-Fc with attenuated IL-12Rβ1 binding (STK-026) to preferentially target activated T cells expressing high levels of IL-12 receptors. STK-026 avoids NK cell hyperactivation but supports sustained T cell activation, interferon γ (IFNγ) production, and intratumoral infiltration of tumor-infiltrating lymphocytes (TILs), macrophages, and cytotoxic NK cells. STK-026 induces tumor control in immune-competent mice, with a substantial therapeutic window between efficacy and toxicity. STK-026 also controls "cold" tumors and synergizes with anti-PD-1 treatment. In non-human primates, STK-026 avoids toxicity associated with IL-12 treatment but sustains effector T cell and NK cell activation. In summary, STK-026 provides anti-tumor efficacy without acute toxicity, expanding the therapeutic index of IL-12 treatment.
    Keywords:  CP: Immunology; IL-12; IL-12Rβ1; IL-12Rβ2; NK cells; STK-026; cytokine; immunotherapy; interleukin 12; t cells
    DOI:  https://doi.org/10.1016/j.celrep.2025.116757
  5. J Transl Med. 2025 Dec 23.
       PURPOSE: This study aimed to define spatial immune subtypes of tumor-infiltrating lymphocytes (TILs) using artificial intelligence (AI)-powered analysis and investigate their association with distinct human epidermal growth factor receptor-2 (HER2) expression levels, response to neoadjuvant chemotherapy (NAC), and patient survival outcomes in triple-negative breast cancer (TNBC).
    METHODS: We conducted a multicenter study involving TNBC patients receiving NAC, stratified into HER2-0, HER2-ultralow, and HER2-low subgroups. An AI-based cell classifier was employed to identify TILs (AI-TILs) and extract spatial coordinates from pathology images. Using unsupervised learning of spatial clustering analysis to identify immune subtypes. The performance of the AI model was benchmarked against four experienced pathologists. Associations among TILs spatial subtypes, HER2 status, NAC response including pathologic complete response (pCR) and residual cancer burden (RCB), and survival outcomes were systematically evaluated.
    RESULTS: AI-TILs demonstrated strong agreement with both senior pathologists, and unsupervised clustering revealed two spatial subtypes: the focal hotspot subtype and the diffuse immune subtype, the latter exhibited broader TIL hotspot areas, lower peak densities, higher pCR rates, and significantly prolonged disease-free survival (DFS). RCB-III status was inversely associated with the diffuse immune subtype, suggesting a link between diffuse immune subtype and good NAC response. Postmenopausal status was independently linked to the focal hotspot pattern. Although HER2 expression was associated with TILs spatial subtypes, it was not an independent influencing factor. In the HER2-low subgroup, the diffuse subtype was significantly associated with better pCR and lower RCB scores, whereas no such relationship was observed in HER2-0 or HER2-ultralow groups. Alluvial and standardized residual analysis further validated that focal hotspot/HER2-low tumors were enriched in RCB-III (residual = 3.26), while diffuse/HER2-low tumors were enriched in RCB-0 (residual = 1.30), suggesting divergent trajectories of immune architecture and chemoresponsiveness.
    CONCLUSION: This study distinguished focal hotspot and diffuse immune subtypes of TILs with distinct clinical implications in TNBC. The diffuse immune subtype correlated with enhanced NAC response and favorable prognosis, particularly in HER2-low TNBC. These findings highlighted the spatial immune architecture as a promising biomarker to refine treatment stratification and deepen our understanding of immune heterogeneity in HER2-low TNBC.
    Keywords:  Artificial intelligence; HER2 expression; Neoadjuvant chemotherapy; Residual cancer burden; Spatial immune subtypes; Triple-negative breast cancer; Tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1186/s12967-025-07608-7
  6. Surg Pract Sci. 2025 Dec;23 100320
       Objectives: This study aimed to investigate the tumor microenvironment (TME) of metastatic PDAC, focusing on tumor-infiltrating leukocytes (TILs) and metabolic checkpoint molecules (MCMs).
    Background: The role of TME in primary and metastatic PDAC is not well understood. Furthermore, the role of energy metabolism in metastatic PDAC is unclear. Therefore, this study aimed to explore the TME in primary tumors and metastases of PDAC, and its prognostic role.
    Materials and Methods: We included 26 cases of metastatic PDAC in this study. We performed immunohistochemistry for TILs and MCMs (HIF-1α, GLUT1, and PDHK1) in primary and corresponding metastatic tumor tissues. We quantified stromal TILs and MCMs using a tumor immune stroma (QTiS) algorithm and correlated the data with clinical outcome.
    Results: We found that CD3+, CD8+, and CD20+ TILs were increased in primary tumors compared to metastatic ones. Kaplan-Meier plots revealed that high infiltration of CD20+ and its combinations in primary tumors correlated with better OS in metastatic PDAC patients. We also found that high infiltration of CD8+ TILs in metastatic tumors correlated with better OS, as did the low density of GLUT1 in both PDAC primary and metastatic tumors. A multivariate Cox regression analysis revealed that CD8+ TILs in metastatic tumors and GLUT1 in PDAC primary and metastatic tumors were independent predictors of survival.
    Conclusion: Distribution of TILs in the TMEs of primary and metastases of metastatic PDAC is different. Our results suggest that TILs (CD8+) and MCMs (GLUT1) in tumor stromal areas can predict OS of patients with metastatic PDAC.
    Keywords:  Metabolic checkpoint molecules, pancreatic ductal adenocarcinoma; Tumor microenvironment; Tumor stroma; Tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1016/j.sipas.2025.100320
  7. J Immunother. 2025 Dec 26.
      Current treatment protocols for bladder cancer (BC) do not include additional immune checkpoint targets or biomarkers that can accurately predict treatment response. Hence, it is imperative to identify more inclusive and promising candidate compounds for immune checkpoint therapy. Therefore, we sought to investigate whether Fibrinogen Like 1 (FGL1) could serve as a new immune checkpoint for bladder cancer. Cell lines overexpressing/silencing FGL1 in human BC cells (5637, HT1376) were constructed, and the regulatory effects of FGL1 on BC cells proliferation, apoptosis, and the tumor immune microenvironment were detected using experimental techniques such as western blot, immunohistochemistry, immunofluorescence, and flow cytometry in an in vivo/vitro experimental model. Silencing FGL1 inhibited BC cells proliferation, promoted BC cells apoptosis, and stimulated tumor-infiltrating lymphocytes (TILs) activation and expansion in tumor microenvironment (TME) for antitumor immunity. Meanwhile, silencing FGL1 down-regulated the expression level of Lymphocyte Activating 3 (LAG3) to inhibit tumorigenicity in xenograft tumor models. Targeting the FGL1/LAG3 signaling pathway can stimulate bladder cancer TILs activation and expansion for antitumor immunity in TME, which inhibits tumor proliferation and growth and promotes tumor apoptosis. Therefore, FGL1 can be used as a potential immune checkpoint for the treatment of BC.
    Keywords:  FGL1; LAG3; bladder cancer; immune checkpoint; tumor immune microenvironment
    DOI:  https://doi.org/10.1097/CJI.0000000000000595
  8. Transplant Cell Ther. 2025 Dec 18. pii: S2666-6367(25)01668-9. [Epub ahead of print]
       BACKGROUND: Non-myeloablative lymphodepleting chemotherapy (LDC) is a crucial component of TIL therapy ensuring T cell engraftment and durable anti-tumor activity. High intensity preconditioning with cyclophosphamide (Cy 60 mg/kg daily for 2 days) and fludarabine (Flu 25 mg/m2 daily for 5 days) dosing is typically used. Given toxicity related to LDC, we investigated the safety and efficacy of reduced-dose LDC followed by standard of care lifileucel and high-dose Interleukin-2 (IL-2) in a select cohort of melanoma patients.
    METHODS: Patients received reduced-dose LDC if they met ≥1 inclusion criteria: age ≥70 years, recently treated brain metastases (BM), bowel metastases, bleeding, need for continuous antiplatelet or anticoagulation use. LDC was given as Cy 30 mg/kg daily for 2 days, Flu 25 mg/ m2 daily for 5 days followed by lifileucel infusion on Day 0 and IL-2 (max: 6 doses). Safety was evaluated using CTCAE v5 and efficacy was reported using RECIST v1.1.
    RESULTS: We treated 17 pts, 9 (53%) males, 8 females (47%) median age: 66 years (range: 36-78). Five had been treated for BM, 59% had LDH ≥ upper limit of normal (ULN), and 35% were BRAFv600 mutant. The median number of IL-2 doses administered was 5 (range: 1-6). IL-2 was discontinued due to hypotension requiring vasopressor (n=1), grade 4 hyponatremia (n=1), infection (n=1), atrial fibrillation (n=1), tachycardia (n=1), grade 3 transaminitis (n=2), combination of tachycardia and/or weight gain, hypoxia, rigors (n=3), and patient refusal (n=1). All patients achieved the grade 4 lymphopenia with a median nadir of absolute lymphocyte count of 0.005 k/µL. Most common grade ≥3 treatment-related adverse events were anemia (n=12, 71%), thrombocytopenia (n=8, 47%), and febrile neutropenia (n=10, 59%) with a median absolute neutrophil count recovery of 7 days. No death occurred within 30 days of TIL infusion. Long term adverse events of grade 3 uveitis (n=1) and grade 3 hearing loss (n=1) developed >30 days after lifileucel. Among 16 evaluable patients, the best response was partial response in 7 patients (44%), stable disease in 3 patients (18%), and progressive disease in 6 patients (38%). Median follow-up was 10 months (95% CI: 8.7 - not reached [NR]), median duration of response was NR, median progression free survival was 5.1 months (95% CI: 4.3 - NR), and median overall survival was NR (95% CI: 10 - NR).
    CONCLUSIONS: Reducing Cy allowed successful completion of TIL therapy among high-risk metastatic melanoma patients without an obvious detrimental effect on response rate. This is the first report demonstrating the safety and feasibility of using real-world objective risk stratification to deliver reduced-dose lymphodepletion prior to lifileucel infusion in high-risk patients with metastatic melanoma. These findings warrant further investigation in a prospective trial of high-risk patients with metastatic melanoma.
    Keywords:  Chemotherapy, Cyclophosphamide; Fludarabine; Lymphodepletion; Melanoma; Tumor-infiltrating lymphocyte therapy
    DOI:  https://doi.org/10.1016/j.jtct.2025.12.009
  9. NPJ Breast Cancer. 2025 Dec 24.
      Hormone receptor-positive (HR + )/HER2-negative (HER2 - ) early breast cancers (BCs) are typically considered immunologically cold. However, combining immune checkpoint inhibitors (ICIs) with chemotherapy has shown to improve pathological complete response (pCR) in high-risk patients. Understanding the relationship between immune activation and tumor biology may help identify HR + /HER2 - BC patients most likely to benefit from such combinations. Baseline gene expression data from two neoadjuvant trials (GIADA and LETLOB) including HR + /HER2 - BC patients were analyzed. PAM50 intrinsic subtyping and relevant immune-related gene signatures were calculated. Tumor-infiltrating lymphocytes (TILs) were assessed on baseline samples. Among 109 tumors, PAM50 classified 44% as Luminal-B (LumB), 33% Luminal-A (LumA), 18% Basal-like, and 5% HER2-enriched. TIL levels (available for N = 101) were generally low (median 2; range 0-100), with higher levels in Basal-like BCs (p = 0.008). Basal-like BCs exhibited significantly higher levels of immune-related signatures (CD8 T-cells, Cytotoxic cells, IFN-γ, response to ICI + CT in GeparNuevo) and of PD-1, PD-L1, and PD-L2 genes. No differences were found between LumA and LumB subtypes. TILs and immune signatures showed a significant weak-to-moderate positive correlation with the basal-like signature. HR + /HER2- BCs that display both features of biological aggressiveness and enhanced immunogenicity may represent ideal candidates for the combination of ICI and chemotherapy.
    DOI:  https://doi.org/10.1038/s41523-025-00886-w
  10. Int J Surg. 2025 Dec 23.
       BACKGROUND: Immunotherapy has significantly improved survival outcomes for various cancer types, despite suboptimal efficacy in over 60% of patients. However, identifying reliable predictive biomarkers for patient response is still a focus of ongoing research, as current indicators often lack consistency. This network meta-analysis (NMA) systematically compared the predictive performance of 13 biomarkers across multiple cancer types to identify optimal predictors of immunotherapy efficacy.
    METHODS: We systematically searched PubMed, OVID, Embase, Cochrane Trials, Web of Science, and trial registries (ClinicalTrials.gov, WHO ICTRP) from inception to September 1st, 2025, and conducted a comprehensive NMA evaluating sensitivity, specificity, diagnostic odds ratio (DOR), superiority, and area under the curve (AUC) for 13 biomarkers. These included circulating tumor DNA (ctDNA), programmed cell death ligand 1 (PD-L1; at varying thresholds), tumor mutational burden (TMB), CD8 + tumor-infiltrating lymphocytes (CD8 + TILs), microsatellite instability (MSI), and inflammatory markers such as neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), lactate dehydrogenase (LDH), Lung Immune Prognostic Index (LIPI), and immune-related adverse effects (irAEs). Subgroup analyses were performed for non-small cell lung cancer (NSCLC), melanoma, gastrointestinal (GI) cancer, urothelial carcinoma, and head and neck squamous cell carcinoma (HNSCC). Heterogeneity and publication bias were assessed using I2 statistics and Deeks' funnel plots.
    RESULTS: This analysis included 54,634 patients from 194 clinical studies worldwide, evaluating various predictive biomarkers. PD-L1 expression was the most frequently assessed (n = 212), with different cutoffs (≥1% [n = 110], ≥ 5% [n = 45], ≥ 10% [n = 32], ≥ 50% [n = 25]), followed by TMB (n = 68). Other markers included NLR (n = 20), irAEs (n = 17), MSI (n = 14), CD8 + TILs (n = 14), ctDNA (n = 14), PLR (n = 12), LIPI (n = 11), and LDH (n = 10). Among these, ctDNA demonstrated the highest sensitivity (0.82, 95% CI: 0.72-0.89) and overall discriminative power (DOR = 9.75, 95% CI: 5.20-16.73; AUC = 0.769). PD-L1 demonstrated threshold-dependent performance: ≥ 50% cutoff demonstrated the highest specificity among PD-L1 thresholds (0.78, 95% CI: 0.73-0.81) and diagnostic accuracy (DOR = 2.60, 95% CI: 1.86-3.52; AUC = 0.661) but the lowest sensitivity (0.42, 95% CI: 0.36-0.49). ≥ 10% threshold showed sensitivity of 0.44 (95% CI: 0.38-0.51) with specificity of 0.74 (95% CI: 0.70-0.78; AUC = 0.656). ≥ 5% cutoff demonstrated sensitivity of 0.54 (95% CI: 0.48-0.60) and specificity of 0.66 (95% CI: 0.62-0.70; AUC = 0.631). Conversely, ≥ 1% cutoff achieved the highest sensitivity among PD-L1 thresholds (0.68, 95% CI: 0.65-0.71) at the cost of the lowest specificity (0.48, 95% CI: 0.45-0.51; AUC = 0.601). TMB balanced sensitivity (0.56, 95% CI: 0.50-0.60) and specificity (0.69, 95% CI: 0.65-0.73; AUC = 0.637). MSI status had the highest specificity (0.89, 95% CI: 0.85-0.93; AUC = 0.727) but low sensitivity (0.36, 95% CI: 0.27-0.46), supporting its role in confirmatory testing. CD8 + TILs showed good sensitivity (0.69, 95% CI: 0.58-0.79) but lower specificity (0.59, 95% CI: 0.49-0.67; AUC = 0.632). irAEs displayed relatively higher sensitivity (0.69, 95% CI: 0.60-0.77) with moderate specificity (0.59, 95% CI: 0.50-0.67; AUC = 0.674). Among inflammatory markers, PLR (AUC = 0.623) showed slightly better predictive power than NLR (AUC = 0.613), while LIPI and LDH exhibited the least overall effectiveness (AUC = 0.585 and 0.544, respectively).
    CONCLUSION: Biomarker performance varies by cancer type and clinical context, underscoring the potential for individualized immunotherapy strategies. ctDNA, PD-L1 (high thresholds, as ≥50%), and TMB emerge as leading predictors, while combinations may optimize sensitivity and specificity. Future research should focus on overcoming heterogeneity and standardization challenges to further refine and individualize immunotherapy approaches and target patients who may benefit from immunotherapy.
    Keywords:  PD-L1; ctDNA; immunotherapy; network meta-analysis; precision oncology; predictive biomarkers; tumor mutational burden
    DOI:  https://doi.org/10.1097/JS9.0000000000004588
  11. J Immunother Cancer. 2025 Dec 21. pii: e013533. [Epub ahead of print]13(12):
       BACKGROUND: Stem-like progenitor exhausted CD8+ T cells (TPEX), located within the tumor-draining lymph nodes (TDLNs), are responsible for maintaining tumor-specific responses in cancer. Although cytokines such as interleukin (IL)-15 are known to expand CD8+ T-cell subsets, transforming growth factor (TGF)-β in the TDLN is known to arrest the egress of these TPEX to the tumor microenvironment. We hypothesized that combining IL-15 stimulatory and TGF-β blocking activity would boost antitumor responses mediated by TPEX in the TDLN.
    METHODS: We developed a bifunctional TGF-βRII/IL-15 protein complex (HCW9218) and evaluated its antitumor activity in two murine models of melanoma and breast cancer. Peripheral blood, TDLN and tumor-infiltrating CD8+ T cells were characterized by flow cytometry following a single subcutaneous dose (s.c.) of HCW9218. Transcription profiling of CD8+ T cells in both murine models was performed. Synergistic activity of HCW9218 with immune-checkpoint inhibitors (ICIs) was evaluated. Finally, safety and immune profiling in patients with chemo-refractory/relapsed solid tumors was performed in a Phase 1 dose-escalating trial.
    RESULTS: HCW9218 was capable of localizing to the TDLNs and tumors after s.c. administration, neutralized TGF-β, expanded TPEX in TDLNs, increased chemokine-expressing effectors in peripheral circulation and promoted their infiltration into murine tumors. These data were corroborated in RNA sequencing analysis of TDLNs. ICIs significantly enhanced the effects of HCW9218 on TPEX and synergistically improved HCW9218 antitumor efficacy in melanoma and reduced spontaneous lung metastasis in breast cancer models. In a Phase 1 clinical trial, HCW9218 monotherapy was well-tolerated, reduced serum TGF-β levels, promoted and sustained CD8+ T-cell expansion in peripheral blood and CD8+ T-cell infiltration in tumor biopsies. Stable disease was reported for four of six subjects (67%) with advanced ovarian cancer treated with HCW9218.
    CONCLUSIONS: Our findings demonstrate that combination therapy targeting immune cells critical for antitumor responses and blocking immune-suppressive environment significantly improves antitumor therapeutic efficacy. These findings provide a strong basis for using HCW9218 to enhance the efficacy of ICIs against solid tumors in the clinical setting.
    Keywords:  Immune Checkpoint Inhibitor; Immunotherapy; Ovarian Cancer; T cell
    DOI:  https://doi.org/10.1136/jitc-2025-013533
  12. Clin Transl Oncol. 2025 Dec 23.
       BACKGROUND: CD8+ T cells play a key role in antitumor immunity, but their heterogeneity leads to inconsistent prognostic associations. CCR5 (C-C chemokine receptor type 5) is a key regulator of CD8+ T cell migration into the tumor microenvironment, suggesting that CCR5+CD8+ T cells may have unique antitumor functions and prognostic significance. However, this hypothesis has not yet been validated.
    METHODS: We investigated the role of CCR5⁺CD8⁺ T cells in tumors and their prognostic value in non-small cell lung cancer (NSCLC) through comprehensive analyses including bulk transcriptomic profiling of TCGA and GEO cohorts, single-cell RNA sequencing of 15 NSCLC patient samples, multiplex immunofluorescence staining of tumor tissues, and in vitro tumor cell co-culture killing experiments.
    RESULTS: We identified that high levels of CCR5+CD8+ T cell infiltration were significantly associated with improved overall survival across multiple cohorts and served as an independent prognostic factor. Gene enrichment analysis indicated that CCR5 upregulation is linked to T cell activation and enhanced antitumor immunity. CCR5 expression correlated more strongly with CD8+ T cell infiltration than that of other CCR family members. Functional assays revealed reduced tumor cell lysis following CCR5 antagonist treatment, indicating impaired T cell cytotoxicity. Single-cell analysis further showed that CCR5+CD8+ T cells represent a functionally potent antitumor subset.
    CONCLUSION: CCR5+CD8+ T cells exhibit antitumor activity in NSCLC and could serve as a promising prognostic biomarker and therapeutic target.
    Keywords:  Biomarkers; CCR5; CD8; Heterogeneity; Lymphocytes; Non; Positive T; Prognosis; Small cell lung cancer
    DOI:  https://doi.org/10.1007/s12094-025-04170-y
  13. Cell. 2025 Dec 24. pii: S0092-8674(25)01369-8. [Epub ahead of print]
      Mechanisms of adaptation of regulatory T cells (Tregs) to harsh tumor metabolic microenvironments for suppression of anti-tumor immunity remain largely unclear. Here, using spatial metabolomics and transcriptomics, we show that human hepatocellular carcinoma harbored metabolically heterogeneous subregions characterized by high glutaminolysis and ammonia contents, where Tregs were frequently present but CD8+ and CD4+ effector T cells die. We found Tregs used the urea cycle to detoxify ammonia by upregulating argininosuccinate lyase (ASL); meanwhile, ammonia was also converted to spermine by the FOXP3 transcription factor regulated spermine synthase (SMS). A direct interaction between spermine and PPARγ was verified by X-ray crystallography, leading to comprehensively modulating the transcription of multiple mitochondrial complex proteins to enhance oxidative phosphorylation and immunosuppression of Tregs. Clinically, anti-PD-1-treated dying tumor cells used transdeamination to release ammonia, which reinforced Treg function, leading to immunotherapeutic resistance. Targeting ammonia production to suppress Tregs presents a potential strategy for anti-tumor immunotherapy.
    Keywords:  Tregs; ammonia; cancer immunotherapy; glutaminolysis; metabolic adaptation; polyamine metabolism; urea cycle
    DOI:  https://doi.org/10.1016/j.cell.2025.11.034
  14. J Exp Clin Cancer Res. 2025 Dec 24. 44(1): 324
       BACKGROUND: The limited response rate and substantial interindividual variability in immunotherapy outcomes remain major barriers to improving prognosis in patients with bladder cancer (BCa). As central effectors of antitumor immunity, the extent of CD8 + T cell infiltration into tumors is a key determinant of immunotherapy response. Members of the histone deacetylase (HDAC) family play critical roles in modulating tumor immune evasion and sensitivity to immunotherapy, making HDAC inhibitors of clinical interest.
    METHODS: A retrospective analysis was performed using data from the IMvigor210 clinical trial and follow-up data from patients with locally advanced BCa who received adjuvant immunotherapy at our center, assessing the association between HDAC1-11 expression and immunotherapy response. RNA sequencing, gene set enrichment analysis (GSEA), chromatin immunoprecipitation PCR (ChIP-PCR), co-immunoprecipitation (Co-IP), mass spectrometry, lysine site mutagenesis, RNA immunoprecipitation, and bioinformatics analysis were employed to outline the HDAC7-BTRC-SRSF7-CCL5 pathway. The immunoregulatory function of HDAC7 was evaluated using CD8 + T cell co-culture assays and tumor models in humanized NOG (HuNOG) mice. Virtual screening, MicroScale Thermophoresis (MST), and HDAC activity assays were conducted to identify potential HDAC7 specific inhibitor. The immunosensitizing effect of Pinocembrin on BCa immunotherapy was validated using a C57BL/6 mouse tumor-bearing model.
    RESULTS: Among the HDAC family members, only HDAC7 expression was significantly associated with immunotherapy response. HDAC7 was overexpressed in BCa and correlated with poorer prognosis. Functional assays demonstrated that HDAC7 suppresses CD8 + T cell infiltration, thereby reducing sensitivity to PD-1 antibody treatment. Mechanistically, HDAC7 reduced acetylation at lysine 24 of the splicing regulator SRSF7, enhancing BTRC-mediated ubiquitination and degradation of SRSF7, which promoted the processing and expression of CCL5 mRNA-a chemokine essential for CD8 + T cell recruitment. Furthermore, Pinocembrin was identified as a selective HDAC7 inhibitor that restores CD8 + T cell infiltration and improves immunotherapy efficacy in BCa.
    CONCLUSIONS: HDAC7 represents a promising diagnostic and therapeutic target in BCa immunotherapy. Pinocembrin, as a specific HDAC7 inhibitor, holds potential as a combination therapy agent to improve immunotherapy response in BCa.
    Keywords:  BCa; CD8 + T cell; HDAC7; Immunotherapy; Pinocembrin; Tumor infiltration
    DOI:  https://doi.org/10.1186/s13046-025-03585-3
  15. Front Immunol. 2025 ;16 1710741
       Introduction: Understanding CD8+ tissue-resident memory T cells (TRM) spatial characteristics in hepatocellular carcinoma (HCC) is challenging, and clarifying the spatial feature changes following immunotherapy represents an urgent research gap.
    Methods: This study employs a multi-omics approach to analyze the spatial distribution and intercellular interactions of TRM cells in HCC tissues using radiomics, single-cell sequencing, and multiplex immunofluorescence histochemistry (m-IHC).
    Results: Our results show that the number of CD8+ TRM cells in HCC increases following immunotherapy. Furthermore, after dividing tumor tissues into the tumor core (TC), invasion margin (IM), and normal tissue (N), a increase in CD8+ TRM cells from the IM to the TC can be observed. Consistent with the results of single-cell sequencing analysis, this change in spatial characteristics may be associated with the interactions between CD8+ TRM cells and CD68+ cells.
    Discussion: Immunotherapy can modify the spatial characteristics of CD8+ TRM cells via regulating their crosstalk with other immune cells, and the spatial distribution of CD8+ TRM cells in the HCC tumor microenvironment (TME) correlates with immune checkpoint blockade (ICB) therapeutic efficacy. Clarifying the mechanisms of action of immunotherapeutic drugs and developing a non-invasive radiomics model to predict CD8⁺ TRM cell dynamics will facilitate the clinical management of HCC.
    Keywords:  CD8+TRM cells; hepatocellular carcinoma; immune checkpoint blockade; immunotherapy; tissue-resident memory; tumor-infiltrating immune cells
    DOI:  https://doi.org/10.3389/fimmu.2025.1710741