bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–10–05
28 papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Exp Hematol Oncol. 2025 Sep 30. 14(1): 121
      Tumor-infiltrating lymphocyte (TIL) therapy, a highly promising form of adoptive cell therapy (ACT), has demonstrated success in treating advanced melanoma. Notably, innovative TIL-based monotherapies and combination regimens have provided durable clinical responses and survival benefits for patients with various solid tumors. This article summarizes recent advances in TIL therapy for solid tumors presented at the 2025 ASCO Annual Meeting, highlighting monotherapies such as Lifileucel, LM103, OBX-115, GT101, GT300, GT201, and HS-IT101, as well as combination strategies with the oncolytic adenovirus TILT-123 or pembrolizumab.
    Keywords:  Adoptive cell therapy (ACT); Clinical study; Solid tumors; Tumor-infiltrating lymphocytes (TIL)
    DOI:  https://doi.org/10.1186/s40164-025-00711-x
  2. Med Oncol. 2025 Sep 28. 42(11): 500
      Adaptive cell therapy (ACT) has emerged as a promising immunotherapeutic approach for cancer treatment by using engineered immune cells to recognize and destroy malignant cells. While ACT has shown remarkable success in hematologic malignancies, its application in solid tumors remains limited due to unique challenges such as limited immune cell infiltration, antigen heterogeneity, and the immunosuppressive tumor microenvironment. This review provides an overview of current strategies to enhance the efficacy of ACT in solid tumors, focusing on engineered T cells, including CAR-T, TCR-T, and tumor-infiltrating lymphocytes (TILs). We discuss recent progress in cancer immunotherapy, with a focus on tumor targeting, resistance to immunosuppressive signals, as well as strategies to overcome antigen escape. Moreover, we highlight the role of gene-editing tools such as CRISPR/Cas9 in designing next-generation immune cells with enhanced functionality and safety. By integrating novel engineering techniques and systems biology approaches, ACT holds the potential to become a key component of personalized cancer therapy for solid tumors.
    Keywords:  Adaptive cell therapy; CAR T cells; NK cells; Solid tumors
    DOI:  https://doi.org/10.1007/s12032-025-03067-8
  3. Front Immunol. 2025 ;16 1655176
      Immunotherapy has transformed the landscape of cancer treatment, offering hope to patients who were once considered beyond the reach of effective care. However, its success is restricted to a limited fraction of patients. This discrepancy in response is largely due to the complex and dynamic nature of the tumor immune-microenvironment. At the heart of this complexity is the concept of cancer immunoediting-a dynamic process through which the immune system both sculpts and is shaped by the tumor. This process unfolds in three key stages: Elimination, Equilibrium, and Escape, each representing a shifting balance between immune defenses and tumor adaptation. Central to this interaction are tumor-infiltrating lymphocytes (TILs) and tumor-associated macrophages (TAMs). TILs are frontline defenders in targeting tumor cells, while TAMs can either hinder or facilitate tumor growth based on their polarization. As cancer progresses, immune selection pressure induces phenotypic alterations that promote immune evasion, fostering an environment detrimental to effective immune response. This review explores the role of these immunological components in each phase of immunoediting and their impact on the efficacy or failure of immunotherapy. Gaining deeper insight into these interactions is crucial for developing advanced immunotherapies that reshape tumor microenvironment and expand the reach of immunotherapy to more patients.
    Keywords:  TAM; TIL; immunoediting; immunotherapy; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1655176
  4. J Immunother Cancer. 2025 Oct 02. pii: e012050. [Epub ahead of print]13(10):
       BACKGROUND: Bladder cancer is unique among visceral malignancies in that urine, which can be easily obtained, has prolonged contact with bladder tumors. Urinary biomarkers offer the potential to provide insight into the host and tumor immune microenvironment to guide therapeutic strategies. We evaluated the immune cellular composition of urine (urine-derived lymphocytes (UDLs)) versus tumor (tumor-infiltrating lymphocytes (TILs)).
    METHODS: We employed high-dimensional flow cytometry analyses on immune cells from tumors (TILs), urine (UDLs), and peripheral blood (peripheral blood mononuclear cells) among patients with bladder cancer. We performed multiplexed immunofluorescence (mIF) of matched tumors to provide spatial context to our findings, comparing deep/invasive and superficial/urine-facing regions of matched tumors.
    RESULTS: Our findings suggest that the CD4+ and CD8+ T cell subsets of UDLs characterized by flow cytometry had similar phenotypic profiles to those found in TILs (cell clusters quantified by multidimensional scaling and differentiation states). Results of mIF imaging with a panel of phenotypic and functional T cell markers suggested that UDLs reflected TILs in both superficial and deep tumor sections. We also found sex-dependent patterns in TILs and UDLs, indicating the male bladder cancer tumor microenvironment is enriched in exhausted CD4+ and CD8+ T cells, while the female bladder cancer microenvironment is enriched for activated T cells.
    CONCLUSIONS: Assessment of UDLs opens avenues of non-invasive biomarker development in clinical settings where bladder cancer TILs are hypothesized to predict clinical response. UDLs may also reflect sex-based differences in antitumor immunity.
    Keywords:  T cell; biomarker; bladder cancer; genitourinary cancer; tumor infiltrating lymphocyte - TIL
    DOI:  https://doi.org/10.1136/jitc-2025-012050
  5. NPJ Breast Cancer. 2025 Oct 02. 11(1): 109
      In the randomized, phase 3 KEYNOTE-119 study, overall survival (OS) was not significantly improved with pembrolizumab 200 mg Q3W versus investigator's choice of chemotherapy in participants with previously treated metastatic TNBC. In this exploratory analysis, we evaluated associations of tumor-infiltrating lymphocytes (TILs), T-cell‒inflamed gene expression profile (TcellinfGEP), BRCA1/BRCA2 mutation (BRCAm) status, homologous recombination deficiency (HRD) status, and tumor mutational burden (TMB) with clinical outcomes. TIL level was associated with improved objective response rate (ORR), progression-free survival (PFS), and OS with pembrolizumab but not with chemotherapy or after adjusting for TcellinfGEP. Associations were also identified between TcellinfGEP and improved ORR, PFS, and OS with pembrolizumab. Participants with TMB ≥ 10 mut/Mb showed a trend toward increased benefit with pembrolizumab versus chemotherapy. No association was seen between BRCAm/HRD status and treatment response. These findings suggest a positive association between TILs, TcellinfGEP, and TMB with clinical outcomes in patients with metastatic TNBC receiving pembrolizumab. ClinicalTrials.gov Identifier: NCT02555657 (date of registration: September 18, 2015).
    DOI:  https://doi.org/10.1038/s41523-025-00814-y
  6. Cancer Drug Resist. 2025 ;8 47
      Aim: Resistance to trastuzumab remains a major barrier to cure in early-stage HER2-positive breast cancer (HER2+ BC). We investigated the impact of genomic alterations and tumor-infiltrating lymphocyte (TIL) density on treatment resistance and survival outcomes. Methods: We retrospectively analyzed 315 patients with HER2+ BC who received adjuvant trastuzumab at Ruijin Hospital (2009-2019). Whole-exome sequencing and TIL scoring were performed on surgical specimens, and clinical and pathological data were collected. The Cancer Genome Atlas (TCGA) cohort was used for external validation. Genomic alterations and TIL density were compared between trastuzumab-sensitive and -resistant tumors. Survival analyses were conducted to identify prognostic biomarkers. Results: After a median follow-up of 109.3 months, 67 tumors (21.3%) were trastuzumab-resistant, exhibiting lower TIL density (mean 19.8% vs. 26.3%, P = 0.001), higher mutation frequencies in FLG, MAP1A, BRCA1, PTPRD, PAPPA2, NCOR2, FBXW7, MYH7, and VCAN, and more frequent alterations in the TP53/NOTCH pathways compared with sensitive tumors (all P < 0.05). A 15-gene trastuzumab response-associated gene (TRAG) signature independently predicted poorer disease-free survival (DFS) in both our cohort (HR, 3.57, P < 0.001) and the TCGA cohort (HR, 4.99, P = 0.037). A high copy number alteration burden was associated with worse overall survival (HR, 2.49, P = 0.043), whereas TIL density > 10% was associated with improved DFS (HR, 2.44, P = 0.003). A prognostic model integrating tumor size, nodal status, estrogen receptor status, TILs, and the TRAG signature showed strong discriminatory power (c-index 0.743 in the training set; 0.915 in the validation set). Conclusion: Genomic alterations and reduced TIL density underpin trastuzumab resistance. The novel TRAG signature and integrated prognostic model enhance risk stratification and may guide personalized adjuvant therapy in early-stage HER2+ BC.
    Keywords:  Breast neoplasms; genomic mutation; models; survival; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.20517/cdr.2025.133
  7. Front Immunol. 2025 ;16 1662216
       Background: Colorectal cancer (CRC) remains a formidable threat to health worldwide. Immunotherapy with immune checkpoint inhibitors results in only a minority of CRC patients experiencing long-term progression-free survival, at the expense of significant autoimmune toxicity. Development of new therapeutics to "wake up" the immune system to fight CRC is necessary. Here we investigated for the first time radioimmunotherapy (RIT) directed towards CCR8, a marker of tumor-infiltrating immunosuppressive T-regulatory cells (ti-Tregs) as a method to recover anti-tumor immunity followed by immunotherapy in CRC models.
    Methods: 225Actinium (225Ac)-labeled anti-CCR8 antibody and anti-CTLA-4 immunotherapy were used to assess their potential synergistic effects in syngeneic murine CRC models CT26 and MC38. The safety of all treatments was assessed through complete blood counts and blood chemistry. 225Ac-anti-CCR8 RIT-treated tumors were analyzed immunohistochemically for FoxP3 and CCR8 expression while mechanistic studies of tumor-infiltrating lymphocytes were done by flow cytometry.
    Results: 225Ac-anti-CCR8 RIT alone demonstrated effectiveness in CRC models but dramatic anti-tumor response was observed when it was combined with anti-CTLA-4 immunotherapy. Immunotherapy alone failed to control tumor growth. Tumor immunohistochemistry post 225Ac-anti-CCR8 RIT showed ablation of CCR8+ ti-Tregs while flow cytometry analysis revealed CCR8-specific increased influx of effector CD8+ T cells, M1 macrophages and NK cells in comparison with 225Ac-control antibody.
    Conclusions: These data demonstrate a synergistic effect of anti-aCCR8 RIT with immunotherapy through enhancement of adaptive and innate anti-tumor responses. Further investigation of anti-CCR8 RIT as a potential cancer-agnostic agent and its combinations with other immunotherapy agents such as anti-PD-1, LAG3 or TIGIT is warranted.
    Keywords:  225Ac-radioimmunotherapy; CCR8; colorectal cancer; immune checkpoint inhibitors; regulatory T cells
    DOI:  https://doi.org/10.3389/fimmu.2025.1662216
  8. ESMO Open. 2025 Sep 30. pii: S2059-7029(25)01626-6. [Epub ahead of print]10(10): 105757
       BACKGROUND: Gastrointestinal (GI) cancers (including esophagus, stomach, colon, rectum, pancreas and liver) account for more than one-quarter of all cancer diagnoses and 35% of cancer-related fatalities worldwide. Quantification of tumor-infiltrating lymphocytes (TILs) is a known cancer prognostic marker. In this study, we used computer vision and machine learning [artificial intelligence (AI)] approaches to evaluate the prognostic significance of computational pathology features relating to spatial arrangement and diversity in the appearance of TILs and cancer nuclei across five different types of GI cancers: colon, stomach, pancreas, and rectum adenocarcinoma and liver hepatocellular carcinoma.
    PATIENTS AND METHODS: The study comprises >1700 patients from four different sites. Pathomic features (2236) were extracted from hematoxylin-eosin stained whole slide images and the top 9 features were selected by Least Absolute Shrinkage and Selection Operator (LASSO) Cox model. The top prognostic features identified were related to the spatial relationships between TILs and the closest cancer nuclei and tumor nuclei shape and texture features captured within local cellular clusters.
    RESULTS: Our trained model identified that 'low-risk' patients have significantly better overall survival than those identified as 'high risk' with a hazard ratio (HR) of 2.28 [95% confidence interval (CI) 1.32-3.93, P = 0.0032] in liver hepatocellular carcinoma; an HR of 2.79 (95% CI 1.66-4.68, P = 0.0001) in pancreatic adenocarcinoma; an HR of 5.85 (95% CI 2.53-15.5, P = 0.0002) in rectal adenocarcinoma; an HR of 1.81 (95% CI 1.07-3.07, P = 0.0268) in gastric adenocarcinoma. Across three different external validation sets of colorectal cancer (CRC) patients, our model yielded an HR of 2.32 (95% CI 1.67-3.23, P < 0.0001) in The Cancer Genome Atlas-Colon Adenocarcinoma (TCGA-COAD), an HR of 2.32 (95% CI 1.67-3.23, P < 0.0001) in Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (PLCO)-COAD, and an HR of 3.38 (95% CI 1.99-5.71, P < 0.0001) in the Emory dataset. Multivariable survival analysis showed that our trained model was prognostic independent of stage, age, race, and sex.
    CONCLUSIONS: Our findings suggest that the spatial relationships of TILs and cancer nuclei are prognostic of survival across multiple GI cancer types.
    Keywords:  artificial intelligence; colon adenocarcinoma; digital pathology; gastrointestinal cancer; machine learning
    DOI:  https://doi.org/10.1016/j.esmoop.2025.105757
  9. bioRxiv. 2025 Sep 24. pii: 2025.09.23.678127. [Epub ahead of print]
      Large-scale CRISPR screening in human T cells holds significant promise for identifying genetic modifications that can enhance cellular immunotherapy. However, many genetic regulators of T cell performance in solid tumors may not be readily revealed in vitro. In vivo screening in tumor-bearing mice offers greater physiological relevance, but has historically been limited by low intratumoral T cell recovery. Here, we developed a new model system that achieves significantly higher human T cell recovery from tumors, enabling genome-wide in vivo screens with small numbers of mice. Tumor-infiltrating T cells in this model exhibit hallmarks of dysfunction compared to matched splenic T cells, creating an ideal context for screening for genetic modifiers of T cell activity in the tumor microenvironment. Using this platform, we performed two genome-wide CRISPR knockout screens to identify genes regulating T cell intratumoral abundance and effector function (e.g., IFN-γ production). The intratumoral abundance screen uncovered the P2RY8-Gα13 GPCR signaling pathway as a negative regulator of human T cell infiltration into tumors. The effector function screen identified GNAS (Gαs), a central signaling mediator downstream of multiple GPCRs that sense different suppressive ligands, as a key regulator of T cell dysfunction in tumors. Targeted GNAS knockout rendered T cells resistant to multiple suppressive cues and significantly improved therapeutic performance across diverse solid tumor models. Moreover, combinatorial knockout of P2RY8 (trafficking) and GNAS (effector function) further enhanced overall tumor control, demonstrating that genetic modifications targeting distinct T cell phenotypes can be combined to improve therapeutic potency. This flexible and scalable in vivo screening platform can be adapted to diverse tumor models and pooled CRISPR libraries, enabling future discovery of genetic strategies that equip T cell therapies to overcome barriers imposed by solid tumors.
    DOI:  https://doi.org/10.1101/2025.09.23.678127
  10. NPJ Breast Cancer. 2025 Oct 02. 11(1): 110
      Metaplastic breast cancer (MpBC) treated with standard chemotherapy has low rates of complete pathological response (pCR)(2-23%). In this study, we evaluate the response to neoadjuvant chemo-immunotherapy (NACI) in early-stage MpBC. Thirty-two stage I-III MpBC patients treated with NACI (KEYNOTE-522 regimen) were prospectively enrolled in an institutional rare tumor program. All MpBC were triple negative; most were of chondromyxoid/matrix-producing (12/32, 38%). The majority had stage II (78%) tumors, 12/32 (37.5%) patients completed NACI, 11/32 (34%) progressed during NACI, and in the remaining 9, NACI was discontinued due to side effects. The pCR rate in the entire cohort was 22% (7/32) and it was statistically higher (5/8, 62%) among patients with high ( ≥ 60%) stromal tumor-infiltrating lymphocytes (sTILs) as compared to patients with < 60% sTILs (1/11, 9%). Most patients received adjuvant systemic therapy (capecitabine 16/32, pembrolizumab 20/32). At a median follow-up of 13 months, there were a total of 2 local recurrences, 10 distant recurrences, and 7 deaths. We demonstrated a modest pCR rate in MpBC with the addition of pembrolizumab (22%). Nonetheless, amongst patients with high sTILs, high pCR rates-comparable to those in the KEYNOTE-522 trial-were observed. These findings suggest that sTILs can be used to triage MpBC patients for NACI.
    DOI:  https://doi.org/10.1038/s41523-025-00816-w
  11. Int J Radiat Oncol Biol Phys. 2025 Jul 17. pii: S0360-3016(25)04483-9. [Epub ahead of print]
       PURPOSE: Although clinical elective nodal irradiation (ENI) often uses conventional fractionation, most animal ENI studies employ hypofractionated protocols. This study evaluated the influence of ENI dose fractionation on tumor immunity and antitumor efficacy in murine models.
    METHODS AND MATERIALS: Splenocytes were irradiated (≤20 Gy) to assess radiosensitivity and immune function. ENI was performed on MC38 and SCC7 tumor models, targeting inguinal lymph nodes with 3 schedules: no ENI (0 Gy), conventional fractionated ENI (Conv-ENI; 2 Gy × 8 fractions), and hypofractionated ENI (Hypo-ENI; 9.8 Gy × 1 fraction). Tumors received 2 Gy × 12 fractions. Antitumor effects, tumor-infiltrating lymphocytes, tumor-draining lymph nodes, and lymphocytes in blood were analyzed longitudinally. Lymphocyte trafficking was inhibited using FTY720 duringENI.
    RESULTS: Irradiation (≤3 Gy) reduced lymphocyte viability in a dose-dependent manner; however, it did not impair immune function, whereas 9.8 Gy reduced viability and impaired immune functions. Hypo-ENI showed inferior antitumor effects compared with Conv-ENI across tumor models, with the no-ENI group demonstrating effective antitumor control. In the MC38 model, CD8+tumor-infiltrating lymphocytes were significantly reduced in the Hypo-ENI group on day 1 but not in the Conv-ENI group. CD8+ lymphocytes in tumor-draining lymph nodes were unaffected across groups on day 1 but significantly reduced in the Hypo-ENI group after FTY720 administration. The number of lymphocytes in blood was significantly reduced in the Hypo-ENI group on day 1.
    CONCLUSIONS: The negative impact of conventional fractionated ENI on tumor immunity and antitumor efficacy may be less severe than previously assumed based on studies using hypofractionated protocols. Future studies should consider the possibility that conventional fractionated ENI and hypofractionated ENI may have different effects on the dynamics of lymphocyte trafficking and their antitumor immunity.
    DOI:  https://doi.org/10.1016/j.ijrobp.2025.06.3845
  12. Cell Rep Methods. 2025 Sep 26. pii: S2667-2375(25)00221-8. [Epub ahead of print] 101185
      Cytotoxic CD8+ T cells are essential mediators of immune responses against viral infections and tumors. Upon antigen encounter, antigen-specific CD8+ T cells undergo clonal expansion and produce effector cytokines, processes that require dynamic metabolic adaptation. However, profiling antigen-specific T cells at single-cell resolution remains technically challenging. We present a spectral flow cytometry-based workflow enabling metabolic profiling of antigen-specific CD8+ T cells identified via major histocompatibility complex (MHC) class I tetramers or CD137 upregulation. The approach integrates the analysis of metabolic protein expression to infer pathway activity, uptake of fluorescent probes to measure functional metabolism and metabolite utilization, and assays evaluating cellular energy metabolism. Applied to human and mouse samples, this method defined the metabolic profiles of cytomegalovirus-, SARS-CoV-2-, and tumor-specific CD8+ T cells across distinct activation states and tissues. By detailing each component of the workflow, we provide practical guidance for applying metabolic spectral flow cytometry to dissect disease mechanisms and therapeutic responses.
    Keywords:  CP: immunology; CP: metabolism; T cell; antigen-specific; metabolism; spectral flow cytometry
    DOI:  https://doi.org/10.1016/j.crmeth.2025.101185
  13. bioRxiv. 2025 Apr 14. pii: 2024.04.01.587634. [Epub ahead of print]
      The efficacy of T cell-activating therapies against glioma is limited by an immunosuppressive tumor microenvironment and tumor-induced T cell sequestration. We investigated whether peripherally infused non-antigen specific autologous lymphocytes (ALT) could accumulate in intracranial tumors. We observed that non-specific autologous CD8 + ALT cells can indeed accumulate in this context, despite endogenous T cell sequestration in bone marrow. Rates of intratumoral accumulation were markedly increased when expanding lymphocytes with IL-7 compared to IL-2. Pre-treatment with IL-7 ALT also enhanced the efficacy of multiple tumor-specific and non-tumor-specific T cell-dependent immunotherapies against orthotopic murine and human xenograft gliomas. Mechanistically, we detected increased VLA-4 on mouse and human CD8 + T cells following IL-7 expansion, with increased transcription of genes associated with migratory integrin expression ( CD9) . We also observed that IL-7 increases S1PR1 transcription in human CD8 + T cells, which we have shown to be protective against tumor-induced T cell sequestration. These observations demonstrate that expansion with IL-7 enhances the capacity of ALT to accumulate within intracranial tumors, and that pre-treatment with IL-7 ALT can boost the efficacy of subsequent T cell-activating therapies against glioma. Our findings will inform the development of future clinical trials where ALT pre-treatment can be combined with T cell-activating therapies.
    Brief Summary: T cell immunotherapies are limited by few T cells in glioma. Adoptively transferred lymphocytes expanded with IL-7 exhibit increased VLA-4 expression and accumulate in tumors.
    DOI:  https://doi.org/10.1101/2024.04.01.587634
  14. Cell Rep Med. 2025 Sep 29. pii: S2666-3791(25)00451-3. [Epub ahead of print] 102378
      Chimeric antigen receptor (CAR) T cells have shown promise in hematological cancers but face challenges in solid tumors, partly due to heterogeneous antigen density. Glypican-2 (GPC2) is an oncofetal antigen highly expressed in neuroblastoma and under evaluation in phase 1 clinical trials. Here, we engineer T cells with antibody-T cell receptors (AbTCRs) targeting GPC2. We generate autologous AbTCR T cells using CT3 or humanized CT3 (hCT3) antigen-binding fragments (Fab) linked to γ/δ T cell receptors (TCRs), along with a CD30 co-stimulatory domain. Both CT3 and hCT3 AbTCR T cells show superior antitumor efficacy compared to CT3 CAR T cells, with hCT3 AbTCR T cells inducing significant regression in neuroblastoma with low GPC2 antigen density. Enhanced efficacy is associated with stronger TCR signaling, expansion of stem cell-like memory T cells, and improved CD8+ T cell infiltration. These results highlight the potential of hCT3 AbTCR T cells for neuroblastoma and indicate broad application of AbTCR T cells in solid tumors.
    Keywords:  AbTCR; CAR; CD30; GPC2; antibody humanization; antibody-TCR; cell therapy; chimeric antigen receptor; gamma/delta TCR; glypican-2; monoclonal antibody; neuroblastoma
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102378
  15. Sci Rep. 2025 Sep 29. 15(1): 33369
      Immune response during the progression of premalignant lesions and their molecular subtype to colorectal cancer (CRC) remains unclear. Using gene expression data from 135 normal (NLs), 176 conventional adenomas (AD), 42 serrated polyps (SER), and 2760 CRC samples, we estimated overall immune activity (ImmuneScore) and tumor-infiltrating lymphocyte (TIL) abundance using the xCell tool. We evaluated association of the ImmuneScore and TILs with CRC progression using adjusted multivariable regression models. Immunohistochemistry (IHC) staining for five immunological markers was conducted on NL, early- and late-stage AD, and carcinoma tissues from 75 study participants. The consensus molecular subtypes (CMS) of adenomas and carcinomas were classified using random forest methods, and the association of immune activity with CRC progression was assessed. Immune activity consistently decreased from NLs through premalignant lesions to adenocarcinoma, more prominently in AD than SER (AD vs. NL: odds ratio = 0.86, 95% CI = 0.84‒0.88; SER vs. NL: 0.89, 0.85‒0.93). Similar patterns were observed in B cells, CD4 + effector memory T cells, CD8 + naïve T cells, and CD8 + cytotoxic T cells. IHC staining of these immunological markers verified their roles in CRC progression. Our analysis revealed that CMS3 is a major subtype of AD. Consistently, higher immune activity was observed in premalignant lesions than in CRCs of the CMS3 subtype. This study provides additional insights into alterations in immune response and their important role in CRC premalignant lesion progression and subtypes.
    Keywords:  Adaptive immune response; Colorectal cancer; Conventional adenoma; Premalignant lesions; Serrated polyps
    DOI:  https://doi.org/10.1038/s41598-025-17653-3
  16. Trends Cancer. 2025 Oct 02. pii: S2405-8033(25)00232-8. [Epub ahead of print]
      Stem-like CD8+ T cells - characterized by high-level expression of the transcription factor TCF-1, and known as progenitor exhausted T (Tpex) cells - have emerged as crucial mediators of durable antitumor immunity. These cells demonstrate unique self-renewal capacity, multipotency, and enhanced responsiveness to immune checkpoint blockade therapy. This review synthesizes current understanding of Tpex cell biology, including their defining characteristics, tissue distribution, and functional importance in antitumor immunity. We focus particularly on innovative approaches to preserve and enhance T cell stemness through combination therapies, cytokine signal modulation, epigenetic regulation, tumor microenvironment modification, and microbiota-based interventions. The development of these next-generation immunotherapies targeting T cell stemness represents a transformative frontier in oncology, holding significant promise for improving therapeutic outcomes in cancer patients.
    Keywords:  TCF-1; cancer; gut microbiota; immune checkpoint blockade; progenitor exhausted T cells; stemness
    DOI:  https://doi.org/10.1016/j.trecan.2025.09.004
  17. Cancer Immunol Immunother. 2025 Sep 29. 74(10): 315
      Chimeric antigen receptor T cell (CAR-T) therapy has revolutionized the treatment of hematological malignancies but faces significant challenges in solid tumor therapy. The tumor microenvironment (TME) of solid tumors, including the dense extracellular matrix (ECM) and immunosuppressive factors, creates physical and biochemical barriers that hinder CAR-T cell infiltration, function, and persistence. Despite advances in CAR-T cell engineering, overcoming these TME barriers remains a major obstacle. The ECM in solid tumors restricts CAR-T cell penetration and limits their antitumor efficacy. Recent studies have explored ECM modulation as a strategy to enhance CAR-T therapy. Nattokinase (NKase), a natural fibrinolytic enzyme, has shown potential in degrading ECM components, promoting TME remodeling, and improving therapeutic responses. In this study, we evaluate the therapeutic efficacy of NKase in combination with MSLN-targeted CAR-T cells for solid tumors. We hypothesize that NKase will reduce tumor fibrosis by degrading ECM components, facilitating CAR-T cell infiltration and enhancing their cytotoxic activity. In vitro and in vivo experiments will assess the effects of NKase on CAR-T cell infiltration, activation, memory phenotype maintenance, and antitumor activity. This study aims to optimize CAR-T therapy for solid tumors and supports the clinical potential of NKase as an adjunctive therapeutic agent.
    Keywords:  Antitumor activity; Chimeric antigen receptor T cell (CAR-T) therapy; Extracellular matrix (ECM); Immune infiltration; Immunotherapy; Mesothelin (MSLN); Nattokinase (NKase); Solid tumors; Tumor fibrosis; Tumor microenvironment (TME)
    DOI:  https://doi.org/10.1007/s00262-025-04167-0
  18. Medicine (Baltimore). 2025 Sep 26. 104(39): e44743
       BACKGROUND: The prognostic and predictive role of the interaction of the immune system with the tumor during the disease process has become increasingly important in recent years. T lymphocyte subgroups in peripheral blood can be used to monitor this dynamic interaction. We evaluated the relationship between baseline peripheral T lymphocyte subsets and treatment response (best objective response) in 47 breast cancer patients receiving neoadjuvant chemotherapy.
    METHODS: This study investigated the impact of initial peripheral T lymphocyte subsets on neoadjuvant chemotherapy response in patients with invasive ductal breast. A total of 47 consecutive patients whose peripheral blood samples were able was included in the study group. The patients' T lymphocyte subsets were measured before treatment. The patients were then followed up to assess their treatment response.
    RESULTS: In our study, we found a significant negative relationship between baseline CD3, CD4, and CD8 absolute counts and pathologic complete response. We also found that high CD4/programmed cell death protein 1 expression (OR: 0.212, 95% CI: 0.061-0.735, P = .015) and high CD8/programmed cell death protein 1 expression (OR: 0.250, 95% CI: 0.073-0.858, P = .028) from baseline peripheral T lymphocyte were correlated with poor pathologic complete response.
    CONCLUSIONS: These results suggest that T lymphocyte subsets may be a predictive biomarker for treatment response in patients with breast cancer undergoing neoadjuvant chemotherapy. Further studies are needed to confirm these findings and to identify the optimal T lymphocyte subset for predicting treatment response.
    Keywords:  T lymphocytes; absolute CD4+ lymphocytes; absolute CD8+ lymphocytes; breast cancer; neoadjuvant therapy
    DOI:  https://doi.org/10.1097/MD.0000000000044743
  19. Nat Biomed Eng. 2025 Oct 01.
      Chimeric antigen receptor (CAR)-T cell efficacy in solid tumours is limited due in part to the immunosuppressive tumour microenvironment (TME). To improve antitumour responses, we hypothesized that enabling CAR-T cells to secrete bifunctional fusion proteins consisting of a cytokine modifier such as TGFβtrap, IL-15 or IL-12, combined with an immune checkpoint inhibitor such as αPD-L1, would provide tumour-localized immunomodulation to improve CAR-T cell functionality. Here we engineer CAR-T cells to secrete TGFβtrap, IL-15 or IL-12 molecules fused to αPD-L1 scFv and assess in vitro functionality and in vivo safety and efficacy in prostate and ovarian cancer models. CAR-T cells engineered with αPD-L1-IL-12 are superior in safety and efficacy compared with CAR-T cells alone and those engineered with αPD-L1 fused with TGFβtrap or IL-15. Further, αPD-L1-IL-12 engineered CAR-T cells improve T cell trafficking and tumour infiltration, and localize IFNγ production, TME modulation and antitumour responses, with reduced systemic inflammation-associated toxicities. We believe our αPD-L1-IL-12 engineering strategy presents an opportunity to improve CAR-T cell clinical efficacy and safety across multiple solid tumour types.
    DOI:  https://doi.org/10.1038/s41551-025-01509-2
  20. bioRxiv. 2025 Sep 23. pii: 2025.09.18.676961. [Epub ahead of print]
      The proliferation of many cancer cells is methionine dependent and dietary methionine restriction (MR) has shown anti-tumor effects in a wide variety of immunodeficiency preclinical models. Yet, whether MR exerts an anti-tumor effect in the presence of an immune-competent background remains inconclusive. Accumulating evidence has shown an essential role of methionine in immune cell differentiation and function. Thus, competition for methionine between tumor cells and immune cells in the tumor microenvironment may drive tumor growth and tumor response to therapy. Here, we aim to define the impact of MR on tumor growth and associated immunity. We first assessed the effect of MR in a series of immunocompetent mouse models of melanoma, colorectal cancer, breast cancer, and lung. MR led to a broad tumor inhibition effect across these models and such tumor inhibition was not sex-or genetic background-dependent but appears to be fully or partially immune-dependent. Through flow cytometry analysis, we found a consistent increase in intratumoral activated CD8 + T cells across different tumor models and depletion of CD8 + T cells partially or completely reversed MR-induced tumor inhibition in a model dependent manner. Interestingly in young healthy non-tumor-bearing mice, MR increased spleen CD3 + and CD8 + T cell populations. Metabolomics and RNAseq analysis of spleen-derived CD8 + T cells revealed significant increase in purine metabolism and amino acid metabolism and that are in line with the metabolic feature of activated T cells. Furthermore, MR improved the efficacy of anti-PD1 immune checkpoint blockade. Together, MR primes T cell metabolism for its anti-tumor effect and improves the efficacy of anti-PD1 checkpoint blockade.
    DOI:  https://doi.org/10.1101/2025.09.18.676961
  21. Cancer Immunol Immunother. 2025 Sep 29. 74(10): 319
       BACKGROUND: Metastatic lung carcinoma poses considerable treatment difficulties. It exhibits cellular characteristics that differ from those of early-stage cancer. Immunotherapy demonstrates enhanced hope for patients with advanced and metastatic lung cancer. Tumor-associated macrophages (TAMs) may be a contributing factor that diminishes the effectiveness of immunotherapy. Secreted phosphoprotein 1 (SPP1)+ TAMs are considered to possess immunosuppressive characteristics, as their interaction with CD8+ T lymphocytes results in the exhaustion of these cells.
    METHODS: We analyzed single-cell RNA sequencing datasets of lung cancer metastases in order to examine the development of brain metastatic tumors. Furthermore, the cell-cell interactions between SPP1+ TAM cells and CD8+ T cells within primary and brain metastatic cancers were systematically compared. Additionally, we utilized a flow cytometer and immunofluorescence to demonstrate how SPP1 affects the function of CD8+ T cells. In vitro, we generated SPP1-overexpressing macrophages and performed qPCR, Western blot, and co-culture assays with or without anti-SPP1 or anti-A2AR treatment to evaluate immunosuppressive effects.
    RESULTS: The results demonstrated that the proportions of immune cells in metastatic brain tissues are lower, while the infiltration of macrophages is higher. SPP1+ TAMs contribute to immune suppression in lung cancer by limiting the activation of CD8+ T cells and cytokine production. Anti-SPP1 treatment positively impacts CD8+ T cell function, counteracting SPP1-induced dysfunction and facilitating the production of cytokines. SPP1 overexpression in macrophages enhanced their immunosuppressive phenotype by upregulating CD73 and cytokines such as IL-10 and TGF-β, leading to impaired CD8⁺ T cell function via A2AR signaling; notably, neutralization of SPP1 or A2AR successfully restored CD8⁺ T cell activity.
    CONCLUSIONS: Our findings characterize the immunological environment of both primary and metastatic lung cancer, highlighting SPP1-mediated immune suppression as a potential therapeutic target to restore T cell responses. Preclinical data demonstrate that Anti-SPP1 antibodies can reverse T cell exhaustion and enhance immune responses in lung cancer.
    Keywords:  A2AR; Brain metastasis; CD73; CD8+ T cell dysfunction; SPP1; SPP1+ TAMs
    DOI:  https://doi.org/10.1007/s00262-025-04180-3
  22. Adv Sci (Weinh). 2025 Sep 30. e11152
      Immune checkpoint inhibitors (ICIs) combined with anti-angiogenic agents manifest improved survival in advanced hepatocellular carcinoma (HCC), but responses remain heterogeneous. Although high PIVKA-II expression correlates with advanced disease stage, early recurrence, shorter survival, and may predict resistance to anti-PD-1 plus lenvatinib therapy, the tumor microenvironment (TME) and resistance mechanisms in HCC with high PIVKA-II expression remain unclear. Clinical data from 156 resected HCC patients and 104 patients treated with anti-PD-1 plus lenvatinib are analyzed to correlate PIVKA-II expression with clinical features and outcomes. Single-cell RNA sequencing (scRNA-seq) is performed on tumors from 15 untreated and 7 treated patients. Mechanistic findings are validated in vitro and in vivo. High PIVKA-II expression is associated with advanced disease stage, increased microvascular invasion (MVI), early recurrence, and poor response to therapy. ScRNA-seq revealed an immunosuppressive TME enriched with regulatory T cells (Tregs), exhausted CD8⁺ T cells, and SPP1⁺ tumor-associated macrophages (TAMs). Mechanistically, tumors with high PIVKA-II expression upregulated NQO1, which stabilized p65 by inhibiting ubiquitination, activating the NF-κB/CXCL12 axis, and recruiting Tregs. This pathway mediated therapeutic resistance. Plerixafor, a CXCL12 inhibitor, disrupted this axis and significantly enhanced anti-tumor efficacy when combined with anti-PD-1 plus lenvatinib in vivo. PIVKA-II is a potentially effective biomarker for predicting resistance to anti-PD-1 plus lenvatinib therapy. Its high expression denotes an immunosuppressive TME. Targeting the NQO1/CXCL12/Tregs axis with Plerixafor may restore sensitivity and improve outcomes.
    Keywords:  CXCL12; Hepatocellular carcinoma; NF‐κB; NQO1; PIVKA‐II; Plerixafor; Tregs; anti‐PD‐1 plus lenvatinib therapy; resistance
    DOI:  https://doi.org/10.1002/advs.202511152
  23. Immunology. 2025 Oct 02.
      Ankylosing spondylitis (AS) is a chronic immune-mediated disease marked by sustained joint inflammation and aberrant bone remodelling. Although chronic antigen exposure usually enforces terminal exhaustion, emerging evidence indicates that a subset of CD8+ T cells in AS evades canonical exhaustion programmes while expressing programmed cell death protein 1 (PD-1). These exhaustion-resistant cells retain effector function and likely contribute to persistent tissue inflammation and structural damage. In this review, we dissect the cellular and molecular basis of exhaustion resistance in AS CD8+ T cells and focus on the convergence of intermittent T cell receptor (TCR) stimulation, metabolic adaptation that preserves mitochondrial fitness, and co-stimulatory inputs from interleukin-15 (IL-15) and CD28. We propose an integrated three-axis model governing CD8+ T cell fate and functional persistence in the AS context shaped by human leukocyte antigen-B27 (HLA-B27) and the gut-joint axis. Clarifying these mechanisms refines current views of T cell dysfunction in chronic inflammation and highlights therapeutic strategies aimed at reprogramming pathogenic immunity in AS.
    Keywords:  CD8+ T cells; HLA‐B27; IL‐15; T‐cell exhaustionankylosing spondylitis; co‐stimulation; exhaustion resistance; gut–joint axis
    DOI:  https://doi.org/10.1111/imm.70044
  24. Int J Surg Case Rep. 2025 Sep 21. pii: S2210-2612(25)01156-3. [Epub ahead of print]136 111970
       INTRODUCTION: Lymph node metastasis is the most important prognostic factor for gastric cancer (GC). While the pN staging system is widely used, it does not account for the total number of dissected lymph nodes, potentially leading to stage migration in patients with suboptimal lymphadenectomy. Alternative systems such as the lymph node ratio (LNR) and log odds of positive nodes (LODDS) may provide superior prognostic accuracy. Our aim is to compare the prognostic significance of the lymph node ratio (LNR), log odds of positive nodes (LODDS), and number of positive lymph nodes (pN).
    METHODS: Eighty-six GC patients treated with curative laparoscopic surgery were retrospectively analyzed. Survival outcomes were assessed using Kaplan-Meier analysis and the log-rank test. Prognostic accuracy was evaluated using receiver operating characteristic (ROC) curves and the area under the curve (AUC) values.
    RESULTS: All three lymph node classification systems, pN, LNR, and LODDS, were significant prognostic factors for survival in gastric cancer, (p = 0.007, 0.002, and 0.036, respectively) based on the log-rank test. Notably, in cases with fewer than 15 lymph nodes dissected, only the LNR system retained prognostic significance (p = 0.037), whereas both LNR and LODDS were effective in the ≥15 lymph node subgroup.
    CONCLUSIONS: LNR and LODDS can be recommended for evaluating lymph node metastasis in gastric cancer, particularly in patients with inadequate lymph node dissection. This is the first study in Vietnam to evaluate and support the integration of LNR and LODDS as complementary prognostic tools in gastric cancer staging.
    Keywords:  Case series; Gastric cancer; Log odds of positive nodes; Lymph node ratio
    DOI:  https://doi.org/10.1016/j.ijscr.2025.111970
  25. Clin Cancer Res. 2025 Sep 29.
      Claudin 18.2 has emerged as a validated target in solid tumors, spurring next-generation therapies. Bispecific antibodies linking Claudin 18.2 to conditional 4-1BB activation offer a novel approach to enhance T cell function. By confining co-stimulation to the tumor microenvironment, they aim to boost efficacy while limiting systemic toxicity.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-25-2554
  26. Cancer Immunol Res. 2025 Sep 30.
      Low or absent expression of major histocompatibility complex (MHC) on tumor cells is a presumed mechanism of resistance to immunotherapy, but evidence for this has largely been indirect. Likewise, whether immunotherapy can be effective without tumor MHC expression is also poorly understood. Using genetically-engineered mouse tumor cells expressing the model neoantigen ovalbumin (OVA), we found that MHC class I-deficient tumor cells, but not MHC class I-sufficient tumor cells, grew progressively when injected subcutaneously into syngeneic C57BL/6 mice. However, combination immunotherapy using agonistic anti-CD40 and dual immune checkpoint blockade (ICB) (anti-PD1 and anti-CTLA-4) was equally effective against tumors that did not express the MHC class I H-2Kb allele, MHC class II, or IFN-γ receptor across multiple pancreatic tumor lines (regardless of OVA). Moreover, CD4+ T cells, but not CD8+ T cells or perforin, were necessary to mediate immunotherapeutic responses. We excluded a role for CD4+ T cell-instructed macrophage-mediated tumor cell death but observed reprogramming of MHC class II-expressing stromal cells within the tumor after anti-CD40/ICB treatment. These data indicate that cancer immune surveillance by T cells does not absolutely require tumor-expressed MHC class I nor CD8+ T cells but instead can facilitate a clinically relevant remodeling of endothelial cells, further underscoring tumor-extrinsic roles for CD4+ T cells as mediators of tumor rejection and durable immune memory.
    DOI:  https://doi.org/10.1158/2326-6066.CIR-24-1342
  27. Commun Med (Lond). 2025 Sep 30. 5(1): 407
       BACKGROUND: Pathologic response has been shown to be strongly associated with long-term event-free survival after neoadjuvant ipilimumab plus nivolumab in stage III melanoma. Only a small proportion of patients developed disease recurrence after initial pathologic response, making conclusions with statistically significant data challenging. However, the homogeneity of population of patients with stage III melanoma might augment the ability to identify immune resistance mechanisms.
    METHODS: To test if recurrence could be due to true tumor immune evasion or due to insufficient persistence of the immune pressure, 10/140 patients with pathologic response after neoadjuvant ipilimumab plus nivolumab with disease recurrence were identified within the OpACIN, OpACIN-neo, and PRADO trials.
    RESULTS: Compared to their counterparts without recurrence, clinical characteristics are different regarding sex, age, BRAF mutation status, depth of pathologic response and frequency of immune-related endocrinopathies. Immune activation-related gene expressions are increased at recurrence after major pathologic response (MPR), but not after pathologic partial response (pPR), and TCR diversity nor clonality are different between baseline and recurrence for both MPR and pPR.
    CONCLUSIONS: No genetic changes explaining tumor immune evasion are found. We propose that disease recurrence may potentially be explained by diminishing of the initial therapy-induced immune response, but not due to genetic changes in the tumor cells mediating immune evasion.
    DOI:  https://doi.org/10.1038/s43856-025-01118-9
  28. Immunity. 2025 Oct 02. pii: S1074-7613(25)00415-7. [Epub ahead of print]
      Interleukin 10 (IL-10)-producing CD4+ type-1 regulatory T cells (Tr1) promote immune tolerance during chronic infection, autoimmunity, and transplantation. However, specific Eomesodermin (Eomes)-dependent stages of Tr1 differentiation and function remain unclear. Using preclinical models of bone marrow transplantation (BMT), we demonstrated a Tr1 differentiation trajectory in vivo from Eomes+IL-10- to Eomes+IL-10+ subsets with the acquisition of cytokine, cytolytic, and exhaustion features. The Eomes+CD4+ fraction represented the dominant cytotoxic subset after BMT, mediating graft-versus-leukemia effects while limiting inflammation. In CD19-targeted chimeric antigen receptor (CAR) T cell immunotherapy, Eomes drove the same CD4+ Tr1 phenotype that controlled cytolysis, while mitigating immune toxicity and promoting persistence. In individuals with high-grade B cell lymphomas that had long-term disease control after receiving commercial CD19-targeted CAR T cells, Eomes+ Tr1 cells represented a stable population comprising 40%-80% of the CD4+ CAR T cell population. Hence, Eomes controls both regulatory and cytotoxic programs in CD4+ T cells, essential for curative immunotherapy outcomes.
    Keywords:  CD4 cytotoxic T lymphocytes; Eomesodermin; bone marrow transplantation; chimeric antigen receptor T cells; cytokine release syndrome; graft-versus-host disease; graft-versus-leukemia; perforin; regulatory T cells; type-1 regulatory T cells
    DOI:  https://doi.org/10.1016/j.immuni.2025.09.004