bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–12–14
28 papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Cureus. 2025 Nov;17(11): e96266
      Introduction Invasive breast carcinoma of no special type (IBC-NST) remains a significant global health burden. Tumor-infiltrating lymphocytes (TILs) reflect the host's anti-tumor immunity and are associated with favorable outcomes, whereas tumor budding (TB) is a marker of tumor aggressiveness. This study aimed to assess TILs and TB in IBC-NST and correlate them with clinicopathological parameters. Methods This retrospective study included 90 (100%) cases of IBC-NST. Stromal TILs (sTILs) and intratumoral TILs (iTILs) were quantified on hematoxylin and eosin (H&E)-stained sections per international guidelines. Tumor buds were counted at the invasive front and classified into low- and high-grade budding. Associations between TILs, TB, and clinicopathological features were analyzed using chi-squared and correlation tests. Results Among 90 cases, 89 (98.8%) were IBC-NST. Both sTILs and iTILs decreased with higher tumor grade, larger size, and increased mitotic count, with statistically significant associations for grade and mitosis (p < 0.05). High-grade TB was significantly associated with lymph node metastasis (35/53, 66.0%), higher tumor grade (50/78, 64.1%), tumor size > 5 cm (23/27, 85.2%), and lymphovascular invasion (32/42, 76.19%) (p < 0.05 for all). A moderate but non-significant positive correlation was noted between sTILs and TB (r = 0.163, p = 0.124). Conclusion Higher TIL levels correlate with favorable tumor characteristics, while increased TB reflects aggressive histological features. A combined evaluation of TILs and TB provides valuable prognostic insights and may assist in stratifying breast cancer patients for tailored therapeutic approaches.
    Keywords:  invasive breast cancer; prognostic marker; tumor budding; tumor microenvironment; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.7759/cureus.96266
  2. Cureus. 2025 Nov;17(11): e96076
       BACKGROUND:  Breast cancer remains a major global health challenge, with tumor-infiltrating lymphocytes (TILs) emerging as a potential biomarker of response to neoadjuvant therapy. This study aimed to evaluate the association between stromal TILs and pathological complete response (pCR) across different molecular subtypes of breast cancer within a South Asian population.
    MATERIALS AND METHODS: A cross-sectional analytical study was conducted at the Department of Oncology, Sindh Medical College, Karachi, from February 15, 2025, to August 15, 2025. A total of 110 female patients with histologically confirmed breast cancer who received standard neoadjuvant therapy and subsequently underwent surgery were included. The cohort comprised all major molecular subtypes (Luminal A, Luminal B, HER2-positive, and triple-negative). HER2-positive patients received anti-HER2-targeted therapy (trastuzumab ± pertuzumab), while triple-negative patients received standard chemotherapy. Stromal TILs were evaluated on pre-treatment biopsy specimens stained with hematoxylin and eosin (H&E), following the International Immuno-Oncology Biomarker Working Group guidelines. A pre-specified cut-off of 20% was used to categorize TILs as low or high, consistent with prior studies and regional validation protocols. Statistical analyses included chi-square tests and multivariate logistic regression to identify independent predictors of pCR.
    RESULTS: The mean age of participants was 48.6 ± 10.2 years, and 58 (52.7%) were postmenopausal. Overall, 40 (36.4%) patients achieved pCR. High TILs were observed in 50 (45.5%) cases and were significantly associated with higher pCR rates compared to those with low TILs (28/50, 56.0% vs. 12/60, 20.0%; p < 0.001). In multivariate analysis, high TILs (adjusted OR 5.25; 95% CI 2.10-13.1; p < 0.001), HER2-positive subtype (adjusted OR 2.10; 95% CI 0.90-4.92; p = 0.08), and triple-negative subtype (adjusted OR 1.75; 95% CI 0.65-4.72; p = 0.27) independently predicted higher pCR rates.
    CONCLUSION: High stromal TILs were strongly associated with improved pathological complete response following neoadjuvant therapy, particularly in HER2-positive and triple-negative breast cancer subtypes. Routine TIL assessment offers a simple, cost-effective, and clinically meaningful biomarker for predicting therapeutic response and guiding individualized treatment strategies, especially within resource-limited regional settings.
    Keywords:  breast cancer; immune response; immunotherapy; neoadjuvant therapy; oncology; pathological complete response; predictive biomarkers; treatment outcomes; tumor microenvironment; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.7759/cureus.96076
  3. Front Immunol. 2025 ;16 1699262
       Introduction: The analysis of tumor-infiltrating lymphocytes often requires techniques that expand their numbers, potentially introducing bias. To address this, we performed a detailed analysis of minimally cultured TILs to evaluate whether this approach better preserves their characteristics.
    Methods: The TIL culture method was based solely on tumor tissue with low IL-2 supplementation to minimize artificial alterations. The validity of this approach was confirmed by the correlation between CD3+ T cell percentages in cultures and infiltration patterns observed by immunohistochemistry. Immunophenotyping, cytokine release, and TCR repertoire analysis were used to characterize CD4+ and CD8+ T cell subsets and their molecular features during minimal expansions.
    Results: High TIL infiltration areas did not consistently correspond to an increased presence of any T cell subset; both CD4+ and CD8+ T cells frequently coexisted in these regions. In contrast, low TIL infiltration sections often displayed a higher proportion of CD4+ T cells. An inverse correlation between CD4+ T cell percentages and cytotoxic molecules was observed, indicating reduced cytotoxic activity in low-TIL sections with abundant CD4+ T cells. TCR repertoire analysis revealed differences between T cell subsets: CD4+ T cells were associated with longer TRA CDR3 nt and shorter TRB N(D)N nt lengths, along with lower diversity, while CD8+ T cells did not exhibit significant correlation with any TCR feature.
    Discussion: This study highlights the distinct biological features of CD4⁺ and CD8⁺ TIL populations within the tumor microenvironment that can be preserved using a minimally expanded TIL approach. The observed associations between IHC patterns, T cell subset composition, cytotoxic potential, and TCR repertoire diversity help identify which biopsy regions yield TILs with greater therapeutic potential, thus providing guidance for TIL selection in immunotherapy.
    Keywords:  TCR repertoire; TIL; breast cancer; immunotherapy; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.3389/fimmu.2025.1699262
  4. Auris Nasus Larynx. 2025 Dec 10. pii: S0385-8146(25)00179-8. [Epub ahead of print]53(1): 27-34
       OBJECTIVE: Immunotherapy using immune checkpoint inhibitors is a standard treatment option for many types of malignancies but is not effective for differentiated thyroid cancer (DTC). Regulatory T-cells (Tregs), which are one of the main suppressive factors in the tumor microenvironment, are another important target for immunotherapy. This study investigated the characteristics of Tregs in DTC with the aim of further developing immunotherapy.
    METHODS: Peripheral blood lymphocytes (PBLs) and tumor-infiltrating lymphocytes (TILs) were obtained from 12 patients with DTC and 12 patients with goiter (benign control group) who underwent primary surgery between February 2017 and September 2022. Flow cytometry analyses were performed for CD4, CD45RA, FOXP3, CC chemokine receptor 4 (CCR4; a candidate Treg-targeting molecule), and immune checkpoint molecules in order to characterize the features of Tregs.
    RESULTS: In the DTC patients, age ranged from 39 to 84 years, and the male:female ratio was 7:5. Pathological staging was pT1 or pT2 in 4 patients, pT3 in 8, pN0 in 5, and pN1 in 7, and extrathyroidal extension was observed in 5 patients. Effector Treg (eTreg) frequency in CD4+T-cells in TILs and PBLs was significantly higher in DTC than in goiter. Extrathyroidal extension was associated with a higher eTreg frequency in TILs. A positive correlation was found between eTreg frequency and the expression of immune checkpoint molecules (PD-1, TIM3, GITR, and OX40) on eTregs in TILs from DTC patients. These findings suggest that Tregs are activated in DTC and are important in the creation of an immunosuppressive microenvironment. In addition, the mean positive rate (±standard deviation) of CCR4 on eTregs was high in PBLs (95.8 % ± 3.8 %) and TILs (92.8 % ± 9.8 %) from DTC patients, suggesting that CCR4 is a potential target for eTreg-depletion immunotherapy for DTC.
    CONCLUSION: Tregs were increased and activated in DTC tumor tissue, indicating that they play an important role in creating an immunosuppressive microenvironment in DTC. The results suggest that eTreg-depletion immunotherapy using an anti-CCR4 antibody (mogamulizumab) might be effective for treating DTC.
    Keywords:  C-C chemokine receptor 4; Immune checkpoint; Immunosuppressive microenvironment; Immunotherapy; Regulatory T-cell; Thyroid cancer
    DOI:  https://doi.org/10.1016/j.anl.2025.12.004
  5. Cancers (Basel). 2025 Nov 24. pii: 3754. [Epub ahead of print]17(23):
      Tertiary lymphoid structures (TLS) arise from lymphoid neogenesis in non-lymphoid organs and are driven by persistent inflammation mediated by chemokines and cytokines. In cancer, TLS orchestrate immune mechanisms relevant to tumor growth and thereby influence clinical outcomes. Observations of a correlation between the presence of TLS and clinical benefits in cancer patients, which suggests that TLS may serve as prognostic and predictive biomarkers, have prompted increased investigation of TLS in tumors. Accumulating evidence indicates that the prognostic and predictive value of TLS is context-dependent and relevant to their cellular composition and functional state. Combining assessments of tumor-infiltrating lymphocytes (TILs) with TLS features yields a more refined prognostic tool by capturing both local immune activity and aspects of the broader tumor immune microenvironment. Improved understanding of TLS biology and of their interactions with TILs can enhance predictions of therapeutic response. Furthermore, therapeutic modulation of TLS composition or function to favor antitumor immunity represents a promising strategy to improve treatment outcomes.
    Keywords:  antitumor immunity; chemokines; immune checkpoint inhibitors; immunotherapy; tertiary lymphoid structures; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.3390/cancers17233754
  6. Comput Struct Biotechnol J. 2025 ;27 5281-5288
       Background: Emerging evidence indicates that microRNAs (miRNAs) play crucial roles in regulating immune checkpoint (IC) genes, thereby modulating IC-related pathways and the behavior of tumor-infiltrating lymphocytes (TILs), thus showing significant potential as a treatment modality in combination therapies with IC inhibitors. Although previous studies primarily focused on single cancer types or individual miRNAs, there is a lack of comprehensive research exploring the complex interactions between miRNAs, IC pathways, and TILs in multiple cancer types.
    Methods: To address this gap, we developed a database called ImmiR that systematically integrates curated data from scientific literature and bioinformatics analyses and defines IC pathways and their ligands/receptors. Using integrated bioinformatics approaches, ImmiR investigated the interactions between miRNAs and IC genes, as well as the associations between miRNAs and TILs.
    Results: ImmiR identified miRNAs targeting genes across 32 immune checkpoint pathways and found associations with 84 immune cell types. As a user-friendly web-based platform, ImmiR provides functional visualizations that facilitate the identification of significant associations from various perspectives such as miRNAs, IC genes/pathways, and cancer types. The database is freely accessible at https://immir.bioinfomics.org/.
    Conclusions: By filling gaps in data resources, ImmiR serves as a vital tool for discovering novel therapeutic targets, refining miRNA-based immunotherapies, and advancing precision medicine in cancer immunotherapy.
    Keywords:  Biomarker; Database; Immune checkpoint; MicroRNA; Tumor-Infiltrating Lymphocyte
    DOI:  https://doi.org/10.1016/j.csbj.2025.11.019
  7. Front Immunol. 2025 ;16 1711415
      Triple-negative breast cancer (TNBC) remains a clinically aggressive subtype of breast cancer, defined by the absence of estrogen receptor, progesterone receptor, and HER2 amplification, and disproportionately affecting younger and racially diverse populations. Despite conventional chemotherapy, TNBC patients often face poor prognoses due to the lack of actionable molecular targets and early metastatic potential. Advances in molecular profiling have unveiled distinct TNBC subtypes and actionable vulnerabilities, including BRCA1/2 mutations and PI3K/AKT/mTOR dysregulation. Therapies targeting DNA repair pathways, angiogenesis, and androgen receptor signaling-particularly via PARP inhibitors and antibody-drug conjugates like sacituzumab govitecan-have demonstrated clinical benefit. Concurrently, TNBC's immunogenic nature, reflected in dense tumor-infiltrating lymphocytes (TILs), has driven the integration of immune checkpoint inhibitors. However, both primary and acquired resistance remain major barriers. This review delineates recent developments in targeted and immunotherapeutic strategies, emphasizing the role of TILs in shaping treatment response and highlighting combinatorial approaches that synergize molecular targeting with immunomodulation. Through a comprehensive understanding of TNBC's molecular and immune landscape, we propose new therapeutic trajectories to improve clinical outcomes in this challenging malignancy.
    Keywords:  antibody-drug conjugates; antitumor immunity; immunotherapy; triple-negative breast cancer; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.3389/fimmu.2025.1711415
  8. Mater Today Bio. 2025 Dec;35 102565
      Programmed cell death protein 1 (PD-1) inhibitors are integral to contemporary cancer immunotherapy. However, their objective response rate in microsatellite-stable colorectal cancer (MSS CRC) remains below 5 %. This limited efficacy is primarily due to insufficient tumor-infiltrating lymphocytes (TILs), a consequence of the combined effects of low tumor immunogenicity and impaired dendritic cell (DC)-mediated antigen presentation. The combination of ursolic acid (Ua) and ginsenoside Rg3 (Rg3) enhances TIL infiltration by inducing immunogenic cell death (ICD) in tumor cells and promoting DC maturation. Due to their poor oral bioavailability, an oral ZIF nanoplatform was engineered with a chitosan oligosaccharide (Cos) and inulin (In) bilayer coating [(In/Cos)@Ua-Rg3-ZIF] for targeted delivery of Ua and Rg3 to the CRC microenvironment. Efficient dual-drug co-loading was achieved with a loading efficiency of 24.07 wt%. After oral administration, the (In/Cos) coating protects the formulation from degradation in the upper gastrointestinal tract, while specific enzymatic cleavage by colonic microbiota releases Ua-Rg3-ZIF into tumor tissues. The ZIF platform responds to elevated adenosine triphosphate (ATP) in the tumor microenvironment, enabling controlled release of Ua and Rg3 to enhance cytotoxic T lymphocyte (CTL) levels. In vivo studies demonstrate that combining (In/Cos)@Ua-Rg3-ZIF with α-PD-1 results in an 80.86 % tumor inhibition rate, significantly outperforming monotherapies. This strategy presents an innovative oral delivery system capable of resisting gastrointestinal degradation and facilitating microbiota-responsive drug release, effectively enhancing antitumor immunity and improving the therapeutic outcome of immunotherapy in MSS CRC.
    Keywords:  Antigen transport; Double-layer glycans; Ginsenoside Rg3; Gut microbiota-responsive ZIF; ICD; Microsatellite-stable colorectal cancer; PD-1 inhibitors; Ursolic acid
    DOI:  https://doi.org/10.1016/j.mtbio.2025.102565
  9. Cell Rep. 2025 Dec 11. pii: S2211-1247(25)01452-4. [Epub ahead of print]44(12): 116680
      Immunotherapy has revolutionized cancer treatment, but it lacks efficacy in a sizable fraction of patients. Therefore, understanding the transcriptional networks that limit CD8+ T cell anti-tumor responses is fundamental. Here, we show that the transcription factor Helios is induced in tumor-infiltrating CD8+ T cells in human and murine cancer. Genetic deletion of Helios in CD8+ T cells reduces tumor growth, decreases the number of intratumoral terminally exhausted CD8+ T cells, and increases the frequency of cells with a transcriptional profile indicative of progenitor capacity. These changes are associated with increased chromatin accessibility in loci encoding stemness-related genes. The combination of Helios and PD-1 deficiencies robustly improves the anti-tumoral capacity of CD8+ T cells. A Helios inhibitor, identified in a small-molecule screen, improves the anti-tumoral effects of PD-1 deficiency. These results demonstrate that Helios represents a therapeutic target that can boost anti-cancer immunotherapy.
    Keywords:  BNTX; CD8(+) T cells; CP: cancer; CP: immunology; Helios; IKZF2; PD-1; T cell exhaustion; TILs; immunotherapy; tumor immunity; tumor-infiltrating T cells
    DOI:  https://doi.org/10.1016/j.celrep.2025.116680
  10. Exp Mol Pathol. 2025 Dec 11. pii: S0014-4800(25)00066-8. [Epub ahead of print]145 105016
      Immunotherapy by immune checkpoint inhibitors (ICIs) revolutionized the treatment of melanoma patients. Tumor-infiltrating immune cells (TIICs) play a crucial role in antitumor immunity activated by ICIs. However, ICIs treatment may be associated with serious immune-related adverse events (irAEs). The aim of the study was to identify the key immune cells and molecules of the tumor microenvironment responsible for the treatment effects and risk of irAEs through immunohistochemical quantification of TIICs. We analyzed metastases (FFPE) of 28 melanoma patients treated with ICIs. Multilevel sampling and stereological quantification were used to assess TIICs identified immunohistochemically by the markers CD1a, CD1d, CD3, CD4, CD8, CD20, CD56, CD68, FOXP3, including immune checkpoint molecules LAG3, PD1, PD-L1. In lymph node metastases, higher infiltration of PD-L1, CD8-positive cells and lower infiltration of CD1a-positive cells predicted response to ICIs (P ≤ 0.05, P ≤ 0.05, P ≤ 0.05, resp.). In all metastasis's subtypes, higher expression of PD-L1 was predictor of response to immunotherapy (P ≤ 0.05). Lower PD-L1 expression (P ≤ 0.05) and lower CD3 expression (P ≤ 0.001) were associated with irAEs. Higher infiltration of CD8-positive T lymphocytes was associated with longer progression-free survival (P = 0.0166) as well as overall survival (P = 0.0454). Stereological quantification of specific immune cells in melanoma metastases, such as T-lymphocytes (CD3), cytotoxic T-lymphocytes (CD8), dendritic cells (CD1a) and PD-L1-positive cells, may predict ICIs treatment efficacy or the risk of irAEs. High infiltration of metastatic tissue by CD8-positive T cells is important for long-term favorable therapeutic response to ICIs.
    Keywords:  Immune-related adverse events; Immunotherapy; Melanoma; Stereology; Tumor infiltrating immune cells; Tumor infiltrating lymphocytes
    DOI:  https://doi.org/10.1016/j.yexmp.2025.105016
  11. JHEP Rep. 2025 Dec;7(12): 101614
       Background & Aims: Atezolizumab plus bevacizumab (Atez/Bev) improves prognosis in advanced hepatocellular carcinoma, but its mechanisms remain unclear. This study aims to identify predictive biomarkers through a comprehensive analysis of the tumor microenvironment.
    Methods: Biopsy samples from 94 patients with advanced hepatocellular carcinoma before Atez/Bev were analyzed using immunohistochemistry, bulk RNA-sequencing, flow cytometry, and multiplexed imaging. The tumor microenvironment assessment included profiling of CD8+ T cells and effector regulatory T (eTreg) cells. Immune dynamics were examined across baseline, in-treatment, and progression samples from each patient.
    Results: We found favorable progression-free survival to be associated with a high percentage of programmed death-1 (PD-1) positivity in CD8+ T cells but not with CD8+ T-cell density. PD-1 positivity in CD8+ T cells was dichotomized at the median (58%). Building upon PD-1 positivity in CD8+ T cells, predominant and diffuse infiltration of CD8+ T cells within the tumor parenchyma was shown to be associated with improved treatment response. PD-1 positivity in eTreg cells was not associated with prognosis. Finally, we found that Bev, an antivascular endothelial growth factor antibody, suppresses the eTreg-cell activation induced by programmed death-ligand 1 (PD-L1) blockade.
    Conclusions: We demonstrate that immunohistochemistry analysis to determine the localization and distribution of CD8+ T cells within the tumor is a viable means of predicting treatment efficacy for Atez/Bev and provides a valuable framework for future trial design. This is an exploratory analysis with no current consequences in clinical practice. Future studies should confirm the results in an external cohort.
    Impact and implications: This study demonstrates that the localization and distribution pattern of CD8+ T cells within the tumor parenchyma are predictors of atezolizumab plus bevacizumab treatment outcomes in advanced hepatocellular carcinoma. The study further reveals that bevacizumab counteracts the potentially unfavorable influence of programmed death-ligand 1 blockade by suppressing effector regulatory T-cell activation. These findings provide both a valuable guide for determining treatment strategies in routine clinical practice and a foundation for future immunotherapy development for the treatment of advanced hepatocellular carcinoma.
    Clinical trials registration: The study protocol was registered on the University Hospital Medical Information Network Clinical Trials Registry (UMIN-CTR) (UMIN000047701).
    Keywords:  atezolizumab; bevacizumab; hepatocellular carcinoma; immunotherapy; tumor microenvironment; tumor-infiltrating lymphocyte
    DOI:  https://doi.org/10.1016/j.jhepr.2025.101614
  12. Blood Adv. 2025 Dec 10. pii: bloodadvances.2025017573. [Epub ahead of print]
      Regulatory T cells (Tregs) are essential for maintaining immune tolerance but also contribute to immune suppression within the tumor microenvironment (TME), dampening anti-tumor immunity in hematologic and solid tumors. As such, strategies aimed at depleting Tregs or reducing their suppressive activity are of great clinical interest. C-C Chemokine receptor 4 (CCR4) is highly expressed on intratumoral Tregs and mediates Treg migration into the TME. Although current therapies targeting CCR4 using monoclonal antibodies have shown some Treg depletion in clinical trials, their clinical efficacy has been limited. We therefore tested whether chimeric antigen receptor (CAR) T cell therapy could be used to deplete Tregs. We evaluated human-specific CCR4-directed CAR T cells (CCR4-CARTs) previously developed for T cell malignancies and determined whether these CARTs could deplete human Tregs ex vivo and in vivo. In patient-derived malignant pleural effusions and lung cancer tumor digests, CCR4 CARTs almost completely depleted Tregs, plus a small population of CCR4+ CD4+ non-Tregs, while sparing CD8+ T cells. When tested in vivo in humanized mice, a single dose of CCR4 CART led to nearly complete Treg depletion. These findings support the potential of CCR4 CARTs as a selective and effective approach to Treg modulation and warrant further clinical investigation.
    DOI:  https://doi.org/10.1182/bloodadvances.2025017573
  13. Cell Death Dis. 2025 Dec 10.
      Although Chimeric antigen receptor (CAR) T-cell therapy has achieved remarkable success in treating hematopoietic malignancies, its clinical application in solid tumors is profoundly hindered by persistent T-cell exhaustion within the immunosuppressive tumor microenvironment (TME). Here, we identified SATB1-a genome organizer regulating chromatin architecture-as a key suppressor of CAR-T cell exhaustion. In Glypican-3 (GPC3)-targeted CAR-T cells, SATB1 was significantly downregulated in tumor-infiltrating exhausted populations. SATB1 overexpression not only reduced expression of multiple inhibitory receptors (PD-1, CTLA-4, TIM3, and LAG-3) but also promoted a central memory phenotype, enhancing cytokine production and cytotoxicity against hepatocellular carcinoma (HCC) cells in vitro. In vivo, SATB1-engineered CAR-T cells exhibited superior tumor control and promoted survival, accompanied by reduced exhaustion markers in tumor-infiltrating T cells. These functional improvements are consistent with the reported role of SATB1 in modulating T cell exhaustion, positioning it as a multifunctional enhancer of CAR-T cell fitness. Collectively, our study unveils SATB1 as a multifunctional modulator that simultaneously targets exhaustion and memory differentiation, offering a novel strategy to enhance CAR-T efficacy against solid tumors.
    DOI:  https://doi.org/10.1038/s41419-025-08307-3
  14. Cancer Med. 2025 Dec;14(23): e71429
       BACKGROUND: Tissue-resident memory T cells (TRM) are a specialized subset of memory T cells that reside in nonlymphoid tissues and draining lymph nodes, playing a critical role in immune surveillance and tumor immunity. Tumor-infiltrated TRM have emerged as key players in immune checkpoint blockade (ICB) therapy, contributing to early therapeutic responses and tumor control.
    AIMS: This review aims to comprehensively summarize the identification, functional roles, and regulatory mechanisms of TRM in the context of tumor immunity, and to explore innovative strategies for enhancing TRM-mediated anti-tumor responses.
    METHODS: We reviewed recent literature on TRM biology, focusing on their transcriptional regulation, phenotypic characteristics, and interactions within the tumor microenvironment. We also reviewed current strategies for modulating TRM function to improve cancer immunotherapy outcomes.
    RESULTS: TRM can remodel the tumor microenvironment, suppress tumor progression, and enhance ICB efficacy. Their function is tightly regulated by transcription factors and phenotypic molecules. Targeted manipulation of these regulatory mechanisms has shown potential in enhancing TRM/TRM-like cell activity and overcoming resistance to ICB therapy.
    CONCLUSION: TRM represent a promising target for cancer immunotherapy due to their potent anti-tumor functions and responsiveness to ICB. Understanding their biology and regulatory networks opens new avenues for therapeutic intervention.
    Keywords:  cancer; immunotherapy; phenotype markers; tissue‐resident memory T cell; transcription factors
    DOI:  https://doi.org/10.1002/cam4.71429
  15. J Clin Invest. 2025 Dec 09. pii: e196907. [Epub ahead of print]
      Adoptive cell therapy (ACT) relies on durable and functional T cells to mediate tumor clearance. Th9 cells are a metabolically fit CD4+ T cell subset with strong persistence but limited cytotoxicity. Here, we identified endomelipeptide A (EpA), a cyclic peptide isolated from Ganoderma lucidum-associated endophytic fungi, as a potent enhancer of Th9 differentiation. EpA promoted a cytotoxic Th9 phenotype with enhanced mitochondrial function and metabolic fitness. Mechanistically, EpA dually targeted ZAP70 and SREBP1, coupling T cell receptor (TCR) signaling activation with lipid metabolism suppression. EpA-treated Th9 cells mediated robust, CD8+ T cell-dependent tumor control and enhanced the efficacy of human Th9 CAR-T therapy in vivo. These findings establish EpA as a distinct cyclic peptide that reprograms Th9 cells and provides a potential approach to boost ACT efficacy.
    Keywords:  Cancer immunotherapy; Immunology; Oncology; Peptides; T cells
    DOI:  https://doi.org/10.1172/JCI196907
  16. Front Immunol. 2025 ;16 1646404
       Introduction: T cell receptor-engineered T cell therapy has emerged as a promising approach in cancer immunotherapy, leveraging the ability of T cells to recognize tumor antigens presented on major histocompatibility complex molecules, offering a targeted approach for treating cancers. This study advances previous research conducted at the Laboratory of Molecular Immunology at RIFCI, where the full repertoire of HER2/neu-specific TCRs was identified. Specifically, here we are functionally validating a distinct TCR clonotype targeting the KIFGSLAFL peptide of HER2/neu protein presented by the HLA-A*02.
    Methods: We employed an integrated approach combining in vitro cytotoxicity assays, single-cell RNA sequencing via BD Rhapsody, secretome profiling via LegendPlex, and in vivo HER2/neu-expressing xenograft models in SCID mice.
    Results: Anti-HER2/neu TCR-T cells exhibited robust antigen-specific cytotoxicity in vitro, preferentially targeting tumor cells with high HER2/neu expression. Single-cell RNA sequencing revealed a unique double-positive (CD4+CD8+) T cell population emerging upon antigen engagement, characterized by a cytotoxic transcriptome with elevated granzyme B, granulysin, perforin, and TNF-α gene expression. Secretome profiling confirmed significantly enhanced production of effector molecules, including IL-2, granzyme B, TNF-α, and IFN-γ, supporting potent T cell activation and function. In vivo, anti-HER2/neu TCR-T cells achieved sustained and significant suppression of tumor growth in HER2/neu-expressing xenograft models, underscoring their therapeutic potential.
    Discussion: These findings validate the broader utility of the previously identified HER2/neu-specific TCR repertoire and elucidate the molecular mechanisms driving its therapeutic efficacy, demonstrating the potential of TCR-T cells for treating solid tumors through robust cytotoxic activity and the emergence of a favorable CD4+CD8+ T cell population. This study offers critical mechanistic insights, establishing a foundation for advancing TCR-engineered therapies toward clinical use in HER2/neu-positive cancers.
    Keywords:  ErbB2; ScRNA-seq; T cells; TCR-T; TCR-T cells; adoptive cell therapy; her2/neu
    DOI:  https://doi.org/10.3389/fimmu.2025.1646404
  17. Cancer Treat Res Commun. 2025 Nov 29. pii: S2468-2942(25)00189-3. [Epub ahead of print]46 101053
      Ovarian cancer remains one of the leading causes of cancer-related mortality among women worldwide. Although standard treatment regimens, including cytoreductive surgery combined with platinum-based chemotherapy, have achieved certain therapeutic advances, the high recurrence rate and the emergence of platinum resistance continue to represent significant clinical challenges. This highlights the urgent need to explore novel therapeutic strategies.As an emerging modality of immunotherapy, PD-1/PD-L1 inhibitors enhance anti-tumor immune responses by modulating the immune system. Although ovarian cancer has traditionally been regarded as a tumor with low immunogenicity, studies have demonstrated that the presence of tumor-infiltrating lymphocytes and the expression of PD-L1 indicate a potential response to immune checkpoint inhibitors.This review focuses on the application of PD-1/PD-L1 inhibitors in the treatment of ovarian cancer, summarizing their mechanisms of action, clinical research progress, potential combination strategies, and future perspectives. A deeper understanding of the role of immune checkpoint inhibitors in ovarian cancer will contribute to optimizing therapeutic strategies and improving patient outcomes. Despite the potential of PD-1/PD-L1 inhibitors in ovarian cancer treatment, overcoming resistance mechanisms, refining patient selection criteria, and improving safety profiles remain key challenges for clinical application.
    Keywords:  Combination therapy; Immune checkpoint inhibitors; Immunotherapy; Ovarian cancer; PD-1 inhibitors; PD-L1 inhibitors
    DOI:  https://doi.org/10.1016/j.ctarc.2025.101053
  18. EBioMedicine. 2025 Dec 09. pii: S2352-3964(25)00520-1. [Epub ahead of print]123 106070
       BACKGROUND: Immunotherapeutic strategies combining Immune checkpoint blockade (ICB) and therapeutic T-cell vaccination hold promise to enhance HIV-1 remission in people with HIV (PWH). While T-cell vaccination alone has shown limited efficacy, ICB may potentiate vaccine-induced T-cell responses. We investigate the functional impact of ICB on vaccine-induced HIV-1-specific CD8+ T-cell responses using relevant samples from PWH receiving early ART and T-cell vaccination.
    METHODS: We conducted comparative laboratory studies on PD-1 and TIM-3 blockade using biological samples from PWH on early ART and T-cell vaccination (Etvac), or without vaccination (Et) from the BCN01 trial, a phase I non-randomised, multicenter therapeutic HIV-1 vaccine study. Also, we included samples from PWH receiving ART during chronic infection stages (Chro). We assessed and characterised vaccine-induced and HIV-1-specific CD8+ T-cell responses using in vitro peptide stimulation, flow cytometry and multiplex assays. We also investigated correlates of response to ICB.
    FINDINGS: PD-1 blockade significantly increased vaccine-induced HIV-1-specific CD8+ T cell responses in the Etvac group, primarily absent prior vaccination, with no effect from TIM-3 blockade. No effect was found in the Et group for any of the tested conditions, while Chro under PD-1 or PD-1/TIM-3 blockade elicited an increase in HIV-1-specific CD8+ T cells. Differentially, vaccine-induced HIV-1-specific CD8+ T cells in response to PD-1 blockade co-express functional markers, sharing a unique profile of soluble molecules. Furthermore, PD-1 expression on CD8+ T cells correlated with in vitro response to PD-1 blockade in Etvac, suggesting its potential as a biomarker in vaccine studies.
    INTERPRETATION: PD-1 blockade enhances functional vaccine-induced HIV-1-specific CD8+ T-cell responses in PWH during early treatment, supporting further clinical investigation into combining ICB to improve therapeutic T-cell vaccine efficacy in PWH.
    FUNDING: This research was partially supported by grants from the National Health Institute Carlos III (PI17/00164), Merck Sharp & Dohme LLC (MSD LKR 155762), a subsidiary of Merck & Co., Inc., Rahway, NJ, USA and the Catalan Government with the European Social Fund (AGAUR-FI_B 00582 and 2021 SGR 00452).
    Keywords:  CD8(+) T cells; Combinatorial immunotherapies; HIV-1; αPD-1; αTIM-3
    DOI:  https://doi.org/10.1016/j.ebiom.2025.106070
  19. J Clin Invest. 2025 Dec 09. pii: e188458. [Epub ahead of print]
      The role of the tumor immune microenvironment (TIME) in modulating responses to antiestrogen therapy in hormone receptor-positive (HR+) breast cancers remains unclear. We analyzed pre- and on-treatment biopsies from patients with HR+ breast cancer treated with letrozole to induce estrogen deprivation (ED). Stromal tumor-infiltrating lymphocytes, assessed by H&E-staining, and immune-related gene sets, including IFNɣ signaling, measured by RNA sequencing, were increased in ED-resistant tumors. Cyclic immunofluorescence and spatial transcriptomics revealed an abundance of CD8+ T cells and enhanced antigen processing and immune gene signatures in ED-resistant tumors. In this group, the expression of CXCL9, CXCL10, and CXCL11 - chemokine genes involved in CD8+ T cell recruitment - and the CXCR3 receptor were upregulated both before and after letrozole. CXCL11 levels were higher in conditioned media from HR+ breast cancer cells co-cultured with CD8+ T cells. Both recombinant CXCL11 and co-culture with CD8+ T cells promoted MCF7 and T47D cell growth in estrogen-free conditions. Finally, deletion combined with silencing of the CXCL11 receptors CXCR3 and CXCR7 in MCF7 cells impaired proliferation in response to exogenous CXCL11 and to co-culture with CD8+ T cells in estrogen-free conditions. These findings suggest that CD8+ T cell-associated CXCL11 in the TIME modulates the response of HR+ breast cancer cells to estrogen suppression.
    Keywords:  Breast cancer; Chemokines; Drug therapy; Immunology; Oncology
    DOI:  https://doi.org/10.1172/JCI188458
  20. NPJ Precis Oncol. 2025 Dec 11.
      We investigated whether artificial intelligence (AI)-based tumor microenvironment profiling correlates with treatment efficacy in unresectable hepatocellular carcinoma (HCC) patients treated with immune checkpoint inhibitor (ICI) therapies. Spatial distribution of immune/non-immune cells from pretreatment H&E images of 163 patients was retrospectively analyzed using an AI/deep-learning model. High tumor endothelial cell (TEC) density was associated with significantly longer progression-free survival (PFS) in the atezolizumab plus bevacizumab (atezo-bev) cohort (HR 0.51 [0.27-0.97]; p = 0.037) but not in the anti-PD-1 monotherapy cohort (HR 1.02 [0.59-1.77]; p = 0.935). Conversely, inflamed immune phenotype, characterized by high intratumoral TIL densities, predicted longer PFS after anti-PD-1 monotherapy (HR 0.50 [0.25-0.99]; p = 0.042) but not after atezo-bev (HR 0.92 [0.50-1.69]; p = 0.762). Our exploratory analysis using AI/deep-learning model demonstrated high TEC density predicted superior outcomes with atezo-bev, while TIL presence correlated with improved anti-PD-1 monotherapy efficacy in HCC patients, suggesting potential clinical applicability in treatment selection.
    DOI:  https://doi.org/10.1038/s41698-025-01207-x
  21. Lancet Digit Health. 2026 Nov 10. pii: S2589-7500(25)00103-7. [Epub ahead of print] 100921
       BACKGROUND: The density of stromal tumour-infiltrating lymphocytes (TILs) is a prognostic factor for patients with triple-negative breast cancer and reflects their immune response. Computational TIL assessment has the potential to assist pathologists in this labour-intensive task, because it can be quick and reproducible. However, computational TIL assessment models heavily rely on detailed annotations and use complex deep learning pipelines that pose challenges for model iterations and clinical deployment. Here, we propose and validate a fundamentally simpler deep learning-based model that is trained in only 10 min on 100 times fewer pathologist annotations.
    METHODS: We collected whole slide images (WSIs) with TIL scores and clinical data of 2340 patients with breast cancer, including 790 patients with triple-negative breast cancer, from three cohorts in three countries (one each in the USA, UK, and Netherlands) and three randomised clinical trials in the Netherlands. Morphological features were extracted from WSIs using a pathology foundation model. Our model, label-efficient computational stromal TIL assessment (ECTIL), directly regresses the WSI TIL score from these features. We trained ECTIL on a single cohort from The Cancer Genome Atlas (n=356, ECTIL-TCGA), on only triple-negative breast cancer samples from four cohorts (n=400, ECTIL-TNBC), and on all molecular subtypes of five cohorts (n=1964, ECTIL-combined). We computed the concordance between ECTIL and the pathologist using the Pearson's correlation coefficient (r) and computed the area under the receiver operating characteristic curve (AUROC) using the pathologist TIL scores split into the clinically relevant TILs-high (≥30%) and TILs-low (<30%) groups. We also performed multivariate Cox regression analyses on the PARADIGM cohort with complete clinicopathological variables (n=384) to assess hazard ratios for overall survival, independent of clinicopathological factors.
    FINDINGS: ECTIL-TCGA showed concordance with the pathologist over five heterogeneous external cohorts (r=0·54-0·74, AUROC 0·80-0·94). ECTIL-TNBC showed a higher performance than ECTIL-TCGA on the PARADIGM cohort (r 0·64, AUROC 0·83 vs r 0·58, AUROC 0·80), and ECTIL-combined attained the highest concordance on an external test set (r 0·69, AUROC 0·85). Multivariate cox regression analyses indicated that every 10% increase of ECTIL-combined TIL scores was associated with improved overall survival (hazard ratio 0·85, 95% CI 0·77-0·93; p=0·0007), which was independent of clinicopathological variables and similar to the pathologist score (0·86, 0·81-0·92; p<0·0001).
    INTERPRETATION: In conclusion, our study showed that ECTIL could score TILs on haematoxylin and eosin-stained, formalin-fixed, paraffin-embedded WSIs in a single step, attaining high concordance with an expert pathologist. Without using deep learning-based segmentation and detection pipelines, ECTIL attained similar hazard ratios to the pathologist's score in an overall survival analysis independent of clinicopathological variables. In the future, such a computational TIL assessment model could be used to pre-screen patients for prospective de-escalation trials in patients with triple-negative breast cancer or as a tool to assist pathologists and clinicians in the diagnostic investigation of patients with breast cancer. Furthermore, our model is available online under an open-source licence, allowing translational researchers to validate and use ECTIL in future studies in breast or other cancers.
    FUNDING: Dutch Cancer Society; Dutch Ministry of Health, Welfare and Sport; and Health∼Holland, Top Sector Life Sciences & Health.
    DOI:  https://doi.org/10.1016/j.landig.2025.100921
  22. Cancer. 2025 Dec 15. 131(24): e70196
       BACKGROUND: Sex differences are a hallmark of urothelial carcinoma (UC), and profoundly influence disease progression and therapeutic outcomes. As critical effector immune cells with intrinsic sexual dimorphism, the contributions of natural killer (NK) cells to this clinical discrepancy remain incompletely understood.
    METHODS: In this multicohort retrospective study of 885 patients with UC (671 males; 214 females), individuals were stratified into NKhigh and NKlow subgroups on the basis of immunohistochemical or transcriptomic evaluation. The association of NK cells with clinical outcomes and the tumor immune microenvironment (TME) was systematically assessed across sexes. Single-cell RNA sequencing and flow cytometry were deployed to decipher the functional state of NK cells.
    RESULTS: Although NK cell infiltration levels were comparable between sexes, their clinical impact was markedly divergent: high NK infiltration conferred a significant survival advantage and superior response to both chemotherapy and immunotherapy exclusively in male patients. Further immune profiling revealed that NKhigh male patients exhibited a coordinated antitumor microenvironment, characterized by immunotype A features, tertiary lymphoid structure formation, and abundant infiltration of dendritic cells and effector T cells. Mechanistically, male-derived NK cells maintained robust cytotoxic and inflammatory programs, whereas female-derived NK cells were polarized toward a stressed, dysfunctional state with decidual-like characteristics.
    CONCLUSIONS: Altogether, this study establishes NK cells as pivotal mediators of sex differences in UC. The stark contrast in NK cell phenotypes underscores a fundamental sexual dimorphism within the TME, which highlights the urgent need for sex-tailored immunotherapy strategies.
    Keywords:  chemotherapy; gender disparities; immunotherapy; natural killer (NK) cells; urothelial carcinoma
    DOI:  https://doi.org/10.1002/cncr.70196
  23. Front Pharmacol. 2025 ;16 1719479
       Background: Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality worldwide. Although immunotherapy and targeted therapy have improved survival in advanced HCC, outcomes remain heterogeneous. Radiotherapy (RT) may enhance systemic treatment efficacy through local control and immunomodulation. Artificial intelligence (AI) offers opportunities to integrate multimodal data for individualized prognostic assessment.
    Methods: A total of 175 HCC patients were included in this study: 115 in the RT group (RT + immunotherapy + targeted therapy) and 60 in the non-RT group (immunotherapy + targeted therapy). Baseline characteristics were analyzed with chi-square and Mann-Whitney U tests. Overall survival (OS) was compared using the Kaplan-Meier method and log-rank test. Patients were randomly divided into a training cohort and a validation cohort (6:4 ratio). Prognostic factors were identified in the training cohort and incorporated into 101 machine learning (ML) algorithms. Model performance was assessed using the concordance index (C-index), receiver operating characteristic (ROC) curves, and risk score stratification.
    Results: The RT group achieved significantly longer OS than the non-RT group (median OS: 15.4 vs. 8.5 months, P = 0.003). Four variables ("Child," "BCLC stage," "Size," and "Treatment") were identified as prognostic factors. Among 101 ML models, the StepCox (forward) + Ridge model showed the best performance (C-index: 0.68 in training, 0.65 in validation). Time-dependent ROC analysis demonstrated AUC values of 0.72, 0.75, and 0.74 at 1-, 2-, and 3-year OS in the training cohort, and 0.72, 0.75, and 0.73 in the validation cohort, respectively.
    Conclusion: RT significantly improved prognosis in advanced HCC patients treated with immunotherapy and targeted therapy. Among multiple algorithms, the StepCox (forward) + Ridge model achieved superior predictive performance, supporting its potential value in individualized prognostic assessment.
    Keywords:  artificial intelligence; hepatocellular carcinoma; immunoradiotherapy; machine learning model; targeted therapy
    DOI:  https://doi.org/10.3389/fphar.2025.1719479
  24. Transl Cancer Res. 2025 Nov 30. 14(11): 7621-7640
       Background: Lung cancer remains a leading cause of cancer-related mortality, with poor survival outcomes. Despite the widespread use of programmed cell death protein 1 (PD-1) blockade therapy, more than 70% of patients fail to benefit and might even develop adverse inflammatory responses. Clinical observations have shown that patients with high neutrophil levels experience worse outcomes following PD-1 blockade. However, the underlying mechanism remains elusive. This study aimed to investigate the impact of neutrophils on anti-PD-1 therapy in lung cancer.
    Methods: Clinical cohort analysis revealed that peripheral neutrophil levels correlated with survival and metastasis in non-small cell lung cancer (NSCLC) patients treated with PD-1 blockade. In this study, we established a subcutaneous Lewis lung carcinoma (LLC) mouse model, and depleted neutrophils by anti-Ly6G antibody with or without PD-1 blockade. Dynamic changes of peripheral immune cells (CD4+T cells, CD8+ T cells, macrophages, neutrophils, and myeloid-derived cells) in mice were examined by Flow cytometry during treatment, and cytokine levels were measured using Luminex multiplex assays.
    Results: Clinically, patients with high neutrophil counts exhibited significantly shorter progression-free survival (PFS) and overall survival (OS), and were more prone to distant organ metastases. In the mouse model, neutrophil depletion enhanced CD8+ T cell infiltration in tumor tissues and suppressed neutrophil-mediated interleukin (IL)-5, IL-6 and IL-17A inflammation, resulting in the inhibition of tumor growth. However, the combination of neutrophil depletion with PD-1 blockade paradoxically restored inflammatory cytokine production and increased myeloid-derived suppressor cell (MDSCs) infiltration, thereby compromising tumor suppression.
    Conclusions: Our observation shows that high neutrophil levels in cancer patients are associated with poorer prognosis and increased risk of distant metastasis. Depletion of neutrophils reshapes the tumor immune microenvironment, wherein excessive inflammatory cytokines drive resistance to PD-1 antibody therapy.
    Keywords:  CD8+ T cell; Lung cancer; inflammation; neutrophil; programmed cell death protein 1 (PD-1)
    DOI:  https://doi.org/10.21037/tcr-2025-1323
  25. J Vis Exp. 2025 Nov 21.
      CD8+CD103+ regulatory T cells represent a specialized immunosuppressive population with demonstrated therapeutic potential in autoimmune disorders; however, their role in ocular surface inflammation remains unexplored. This protocol describes a systematic approach for isolating CD8+CD103+ T cells from mice exposed to desiccating stress and evaluating their therapeutic efficacy through adoptive transfer in experimental dry eye disease. The methodology encompasses magnetic-activated cell sorting for CD8+CD103+ T-cell purification, standardized desiccating stress induction, and comprehensive functional assessments, including tear production measurement, corneal barrier permeability testing, and molecular analysis of inflammatory mediators. Representative results demonstrate that CD8+CD103+ T cell therapy significantly restored tear production, preserved conjunctival goblet cell populations, and maintained corneal epithelial barrier integrity. Treatment achieved substantial suppression of matrix metalloproteinase expression, reduced apoptotic cell death, and modulated cytokine profiles from pro-inflammatory to tissue-protective patterns. The protocol effectively reduced CD4+ T cell infiltration in ocular tissues, while simultaneously suppressing pathogenic IL-17A and IFN-γ production and enhancing protective IL-13 levels. This approach provides a robust experimental framework for investigating CD8+CD103+ T cell-mediated immunomodulation in ocular surface disorders and establishes a foundational methodology for developing novel cell-based therapeutics for dry eye disease management.
    DOI:  https://doi.org/10.3791/69110
  26. Ann Med Surg (Lond). 2025 Dec;87(12): 9193-9194
      Chimeric antigen receptor (CAR)-T cell therapy, despite revolutionizing hematological malignancies, remains limited in solid tumors due to immunosuppressive microenvironments and systemic toxicities from combination immunotherapies. Recent engineering innovations demonstrate that physically linking anti-PD-L1 antibodies to interleukin-12 within CAR-T cells creates tumor-localized immunomodulation, concentrating therapeutic activity at PD-L1-positive sites while minimizing systemic exposure. In preclinical models, PD-L1-binding IL-12 fusion proteins achieved superior antitumor responses (100 vs. 50% complete responses) compared to non-binding controls, with significantly reduced inflammatory toxicity. Spatial proteomic analysis revealed comprehensive tumor microenvironment remodeling including enhanced CD8+ T cell infiltration and reduced immunosuppressive myeloid populations. Validation in human CAR-T cells targeting TAG72-positive ovarian cancer confirmed appropriate PD-L1 binding and enhanced cytotoxicity. This rational engineering strategy addresses multiple barriers simultaneously through molecular sequestration, offering a promising platform applicable to alternative checkpoint-cytokine combinations and other cellular therapeutics. Clinical translation represents a critical next step for extending CAR-T efficacy to solid malignancies.
    Keywords:  CAR-T cells; checkpoint inhibitors; immunotherapy; solid tumors; tumor microenvironment
    DOI:  https://doi.org/10.1097/MS9.0000000000004256
  27. Hepatobiliary Surg Nutr. 2025 Dec 01. 14(6): 1042-1044
      
    Keywords:  Hepatobiliary cancers; T cell immunotherapy; chimeric antigen receptor T cell therapy (CAR-T therapy); organoid models; tumor microenvironment
    DOI:  https://doi.org/10.21037/hbsn-2025-aw-863