bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–12–21
nineteen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. J Immunother Cancer. 2025 Dec 17. pii: e013373. [Epub ahead of print]13(12):
       BACKGROUND: Adoptive transfer of T-cell receptor-engineered T cells (TCR-T cells) has shown promising efficacy in solid tumor treatment, but achieving clinical benefit typically requires infusion of tens of billions of cells. The commonly used rapid expansion protocol (REP), based on the CD3-agonistic OKT3 antibody and irradiated allogeneic feeder cells, exponentially expands tumor-infiltrating lymphocytes (TILs). However, the effect of REP on TCR-T cell frequency and phenotype remains unclear. This study aimed to evaluate the impact of REP on TCR-T cells and to assess the potential of a neoantigen-specific stimulation platform, NeoExpand, as a strategy to selectively expand TCR-T cells with favorable phenotypes.
    METHODS: We compared the effects of REP and NeoExpand on the frequency, yield, and phenotype of TCR-T cells engineered against shared neoantigens. Various autologous antigen-presenting cell (APC) types, including dendritic cells and bulk peripheral blood mononuclear cells (PBMCs), were tested as stimulators in NeoExpand. Additionally, we combined NeoExpand with CD3 activation, with or without allogeneic feeders, to improve scalability. The role of exogenous interleukin (IL)-21 in shaping TCR-T cell phenotypes was also investigated.
    RESULTS: REP impaired the frequency and phenotype of TCR-T cells, particularly CD4+ and CD8+ subsets matched to their engineered TCRs. In contrast, NeoExpand selectively increased TCR-T cell frequency and improved their phenotypes but did not consistently achieve higher total yields compared with REP. Among the tested autologous APCs, PBMCs effectively supported selective expansion. Further optimization using PBMCs as APCs revealed that combining NeoExpand with CD3 activation enabled exponential expansion without compromising selectivity. Inclusion of IL-21 during NeoExpand promoted a naïve/stem-like phenotype in CD8+ TCR-T cells, with minimal effect on CD4+ TCR-T cells.
    CONCLUSIONS: These findings demonstrate that NeoExpand enables selective expansion of TCR-T cells while preserving favorable phenotypes and scalability. The approach supports further clinical evaluation of NeoExpand as a strategy to generate high-quality TCR-T cell products for adoptive cell therapy.
    Keywords:  Adoptive cell therapy - ACT; Immunotherapy; T cell; T cell Receptor - TCR
    DOI:  https://doi.org/10.1136/jitc-2025-013373
  2. bioRxiv. 2025 Dec 16. pii: 2025.12.10.693486. [Epub ahead of print]
      Cancer immunotherapies, from checkpoint blockade to adoptive cell therapies like tumor-infiltrating lymphocytes (TILs), have revolutionized cancer treatment but are limited by variable efficacy and significant toxicities. A central challenge is identifying ideal T-cell populations that effectively eliminate tumors without causing off-target damage, a distinction not captured by existing biomarkers. We show that co-expression patterns of chemokine receptors (CRs) CXCR3, CCR5, and CXCR6 on CD8 + T cells provide a functional "code" defining subsets with divergent roles in on-target immunity versus off-target inflammation. In mouse and human melanoma, a triple-positive (CXCR3 + CCR5 + CXCR6 + ) T-cell subset is essential for tumor control, and its genetic signature correlates with positive clinical response, while a distinct CCR5 + CXCR6 + subset drives liver immune-related adverse events (IRAEs). Crucially, this CR code reveals that immunotherapy actively reshapes T-cell trafficking patterns, uncovering profound heterogeneity within conventional populations and distinguishing potent anti-tumor progenitors from cells predisposed to exhaustion or off-target migration. This work establishes CR co-expression as a practical tool, providing a surface marker-based strategy to identify and enrich optimized T cells for adoptive therapies, thereby offering a framework to uncouple efficacy from toxicity.
    DOI:  https://doi.org/10.64898/2025.12.10.693486
  3. BMC Cancer. 2025 Dec 15. 25(1): 1870
       BACKGROUND: Inflammatory blood markers and tumor-infiltrating lymphocytes (TILs) are associated with the prognosis of various cancers. However, reports on their value as prognostic markers in soft tissue undifferentiated pleomorphic sarcoma (UPS) are limited. We aimed to clarify the predictive value of these markers for the prognosis of patients with UPS, focusing on resectable tumors of the extremities and trunk.
    METHODS: This retrospective analysis included data from 103 patients with localized UPS in the extremities and trunk. The neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), lymphocyte-monocyte ratio (LMR), and systemic inflammatory response index (SIRI) were calculated, and the median values were determined as cut-off values. Immunohistochemical staining of CD8 + TILs was also performed. Disease-specific overall survival (DOS) and distant metastasis-free survival (DMFS) rates were analyzed using the Kaplan-Meier method, and prognostic factors were identified using the Cox proportional hazards model.
    RESULTS: Among the inflammatory blood markers, SIRI was found to be a sensitive prognostic factor, and a high SIRI was associated with worse DOS (P = 0.16) and DMFS (P = 0.09). Although CD8 + TILs were not associated with DOS (P = 0.67), high CD8 + TILs correlated with improved DMFS (P = 0.20). Only tumor size > 10 cm was significantly associated with worse DMFS (P = 0.02) in the univariate analysis, while inflammatory blood markers and CD8 + TIL showed no correlation. The combination of SIRI and CD8 + TILs revealed that high CD8 + TILs in tumor tissue could improve DMFS (P = 0.04) and DOS (P = 0.15) in patients with high SIRI compared to those with low CD8 + TILs and high SIRI.
    CONCLUSIONS: In patients with localized UPS, CD8 + TILs infiltration into the tumor tissue could improve the prognosis of patients with high SIRI.
    Keywords:  CD8 + tumor-infiltrating lymphocyte; Soft tissue; Systemic inflammatory response index; Undifferentiated pleomorphic sarcoma
    DOI:  https://doi.org/10.1186/s12885-025-15182-w
  4. bioRxiv. 2025 Dec 09. pii: 2025.12.05.692662. [Epub ahead of print]
       Background: Adoptive cell therapy using tumor-infiltrating lymphocytes (TIL) is approved for the treatment of advanced melanoma but is limited by the need for patients to undergo surgical tumor resection. Malignant pleural effusions (MPE) may represent a more accessible source of tumor-reactive T cells. Here, we characterize the cellular composition as well as the transcriptional and functional properties of T cells from MPE compared with pulmonary metastasis and blood from a patient with melanoma.
    Methods: The immune cellular composition was immunophenotyped by high-dimensional flow cytometry from synchronously collected MPE, a lung metastasis, and blood from a patient with metastatic melanoma. Sorted CD3 + T cells were profiled by single-cell RNA sequencing (scRNA-seq) and T cell receptor sequencing (scTCR-seq). TCR reactivity to autologous tumor was evaluated through in vitro activation assays with TCR-transduced Jurkat and autologous cancer cells. The killing capacity of ex vivo expanded T cells of autologous cancer cells was assessed through in vitro cytotoxicity assays.
    Results: MPE had higher proportions of CD45 + immune cells and CD3 + T cells (70.5% vs 50%) compared with tumor and was enriched for effector CD8 + T cells, CCR7 - CD45RA - effector memory CD4 + T cells, and quiescent CD25 high CD127 low regulatory CD4 + T cells. MPE T cells exhibited lower levels of co-inhibitory receptors (PD-1, LAG-3, TIGIT, TIM-3) expression relative to tumor. ScRNA-seq showed enrichment of NK-like effector CD8 + T cells in MPE. Pseudotime analysis indicated that MPE T cells were less exhausted than tumor T cells. The clonal repertoire of MPE and tumor highly overlapped, including 62.2% of predicted neoantigen-specific (NeoTCR) clonotypes. Notably, clonally-related NeoTCR T cells in MPE exhibited higher cytotoxic and stemness, and lower exhaustion signatures compared with sister clones in the tumor. Two of four selected NeoTCR clonotypes transduced in Jurkat cells demonstrated MHC class I-restricted reactivity in co-culture with autologous cancer cells. MPE T cells also readily expanded in the presence of high-dose IL-2 and demonstrated MHC class I-dependent killing of autologous cancer cells.
    Conclusions: MPE harbors polyclonal, tumor-reactive T cells with lower features of terminal exhaustion and higher cytotoxic potential relative to tumor T cells. MPE may therefore serve as a more accessible source for TIL therapy.
    DOI:  https://doi.org/10.64898/2025.12.05.692662
  5. Mol Ther Oncol. 2025 Sep 18. 33(3): 201041
      The disruption of immune checkpoints in T cells is a promising tool for improving the efficacy of tumor-infiltrating lymphocyte (TIL) therapy. While CRISPR-Cas9 genome-editing is efficient, Cas9 nucleases induce double-strand DNA breaks and risks improper translocations, inversions, and chromosomal deletions in engineered T cells. Cas9 nickase (nCas9) used in base-editing cuts only a single strand of DNA, reducing genetic aberrations in modified cells. Here, we established a small-scale, good manufacturing practice-compatible adenine base editing (ABE) procedure for both single and dual knockout of co-inhibitory receptors TIM3 and TIGIT in TILs. ABE-mediated conversion of A·T to G·C pairs in TIM3 and TIGIT specific splice-sites led to high knockout efficiency, with negligible insertion-deletion events post editing. Using melanoma and ovarian TILs, we show that target-specific editing by ABE of TIM3 and TIGIT improved (1) TIL fold-expansion during the rapid expansion protocol without adversely impacting phenotype, (2) cytokine production, and (3) serial killing upon co-culture with autologous patient-derived tumor cells in vitro. Moreover, dual edited TILs were able to infiltrate tumor spheroids in vitro and control patient-derived tumors in vivo. Taken together, we show the feasibility of ABE multiplex editing as a promising tool for engineering TILs for clinical applications.
    Keywords:  ABE; ACT; MT: Regular Issue; TIL; adenine base editing; adoptive cell transfer; cancer immunotherapy; gene editing; tumor-infiltrating lymphocyte
    DOI:  https://doi.org/10.1016/j.omton.2025.201041
  6. Cancer Immunol Immunother. 2025 Dec 19. 75(1): 20
       BACKGROUND: Tumor-infiltrating lymphocytes (TILs) are essential to effective immunotherapy for hepatocellular carcinoma (HCC). Among factors regulating TILs, tumor-associated high endothelial venules (TA-HEVs) have been identified in various cancers and may contribute to TIL recruitment. This study aimed to investigate the clinical significance of TA-HEVs in HCC and their association with TILs.
    METHODS: 156 patients with HCC who underwent hepatic resection were analyzed. TA-HEVs were identified using double immunohistochemical staining for MECA-79 and CD31. Clinical outcomes and TIL density were compared between TA-HEV-present and -absent cases. Multicolor flow cytometry and RNA sequencing of tumor tissues were also performed.
    RESULTS: TA-HEVs were observed within tertiary lymphoid structures (TLSs) surrounding tumors in 31 of the 156 cases. The proportion of TLSs containing TA-HEVs increased with TLS maturation. TA-HEV-present cases had significantly better disease-free survival (3 years: 79.8% in TA-HEV-present cases vs 60.1% in TA-HEV-absent cases, p = 0.011). Multivariate analysis identified TA-HEVs as an independent favorable prognostic factor. Moreover, TA-HEV-present cases had significantly higher densities of intra-tumoral CD4⁺ and CD8⁺ T cells. RNA sequencing revealed enhanced pathways related to immune response and lymphocyte activation in TA-HEV-present cases, with high expression of cytotoxic lymphocyte markers. Flow cytometry confirmed elevated proportions of activated CD8⁺ T cells in TA-HEV-present cases.
    CONCLUSIONS: TA-HEVs in HCC were associated with significantly better prognosis and may contribute to immune activation within the tumor microenvironment.
    Keywords:  Immune checkpoint molecules; Primary liver tumor; Tertiary lymphoid structure; Tumor microenvironment; Tumor-infiltrating lymphocyte
    DOI:  https://doi.org/10.1007/s00262-025-04265-z
  7. Cancer Res. 2025 Dec 15.
      Lipid metabolism reprogramming modulates the tumor microenvironment (TME) and alters the function of immune cells, including tumor-infiltrating lymphocytes (TILs). While lipids can enhance general T cell activity, high lipid content in the TME may restrain the anti-tumor function of effector T cells and augment immunosuppression by regulatory T cells. In addition, lipid metabolism reprogramming greatly influences the crosstalk between TILs and other immune cells in the TME, including dendritic cells, macrophages, and myeloid-derived suppressor cells. By discussing potential therapeutic strategies to target lipid metabolism in TILs, along with combination approaches with chemo-immunotherapy, nanomedicine, and adoptive cell transfer therapy, we aim to lay the groundwork for advancing effective treatments for cancer patients.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-25-2568
  8. Mol Immunol. 2025 Dec 16. pii: S0161-5890(25)00290-1. [Epub ahead of print]189 142-152
       BACKGROUND: Adoptive cell therapy (ACT) utilizing tumor-infiltrating lymphocytes (TIL) is a promising immunotherapeutic approach for disseminated malignancies. However, ex vivo-expanded tumor-infiltrating lymphocytes (TIL) often rapidly progress to a state of functional exhaustion or suppression within the tumor microenvironment. Interleukin-7(IL-7) not only supports the survival of T-lymphocyte in vivo but also induces vigorous expansion of naïve and memory T lymphocytes in vitro. Upregulating the expression of interleukin-7 receptor alpha (IL-7Rα) helps T cells restore their responsiveness to IL-7 signaling, aids in their survival, and enables them to regain anti-tumor activity.
    METHODS: Firstly, we utilized databases to analyze the expression changes of CD127 in different tumor tissues, clarifying the correlation between its expression changes and patient prognosis. Subsequently, we collected clinical patient samples to validate the expression changes of CD127 in tumor-infiltrating T cells. We also simulated the tumor microenvironment through in vitro co-culture to explore the impact of tumor cells on the expression of CD127 on the surface of T cells. After then, we screened for miRNAs that are complementary to the sequence of the TATA box region in the CD127 promoter and employed a CD127 promoter driven dual-luciferase reporter system to identify the specific miRNA capable of upregulating CD127 expression. Finally, we analyzed the effects of miRNA-mediated upregulation of CD127 on T cell function.
    RESULTS: Bioinformatics analysis and clinical validation both confirmed decreased IL-7Rα expression in tumors. Moreover, in clinical samples, IL-7Rα and miR-3188 expression levels showed concordant changes and a positive correlation. miR-3188 can upregulate the expression level of IL-7Rα by specifically targeting the TATA-box region of CD127 promoter. Utilizing miR-3188 to upregulate IL-7Rα expression can facilitate T cell survival, promote the development of memory T cells and enhance the secondary response and tumor-killing capacity of T cells.
    CONCLUSION: Our findings reveal a novel mechanism of IL-7Rα regulation and propose a potential strategy to improve the persistence and functionality of T cells for ACT.
    Keywords:  IL-7Rα; MiR-3188; TIL; Tumor
    DOI:  https://doi.org/10.1016/j.molimm.2025.12.007
  9. Cell Rep Med. 2025 Dec 16. pii: S2666-3791(25)00579-8. [Epub ahead of print]6(12): 102506
      Adoptive cell therapy (ACT) targeting tumor-specific antigens holds promise for solid tumors, but limited neoantigen presentation remains a key barrier to efficacy. Here, we identify and characterize a T cell receptor (TCR), T104, for the KRAS.G12V mutation, a prevalent neoantigen in colorectal, lung, and pancreatic cancers. TCR-T104 selectively recognizes and kills KRAS.G12V-expressing tumor cells. Combining T cell therapy with lymphodepleting chemotherapy significantly enhances tumor cell killing, particularly by TCR-T cells, tumor-infiltrating lymphocytes (TILs), and T cell engager antibodies across multiple cancer types and target antigens. Mechanistically, chemotherapy upregulates immunoproteasome activity and human leukocyte antigen (HLA)-I surface expression. HLA-immunopeptidome analyses reveal that chemotherapy remodels the antigenic landscape across tumor cell lines and in vivo models, increasing peptide abundance and hydrophobicity while altering proteasomal cleavage preferences. These findings establish a synergistic role for chemotherapy in enhancing neoantigen presentation and T cell-mediated tumor recognition and suggest that fine-tuning these regimens could improve ACT efficacy, particularly in tumors with low-abundance neoantigens.
    Keywords:  Adoptive Cell therapy; KRAS G12V; TCR-T therapy; colon cancer; immunopeptidomics; immunoproteasome; lung cancer; lymphodepleting chemotherapy; neoantigens; pancreatic cancer
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102506
  10. Nat Commun. 2025 Dec 15.
      Overcoming barriers in solid tumor immunotherapy remains challenging, with adoptive T cell therapies limited by antigen loss, poor tumor infiltration, and T cell exhaustion. Here, we present a nanoengineered tumor-infiltrating lymphocyte (TIL) therapy using anti-PD-1 antibody (αPD-1)-conjugated ZIF-8 nanoparticles to effectively suppress pancreatic tumor growth. These nanoparticles release hyaluronidase (HAase) and decitabine (DEC) in acidic tumor microenvironments, promoting stroma degradation and C-C motif chemokine ligand 5 (CCL5) secretion, while retaining αPD-1 on TILs to prevent exhaustion. CCL5 recruits additional nanodrug-loaded TILs for further release of HAase and DEC, establishing a self-reinforcing infiltration loop. This approach increases TIL infiltration by 12-fold in immunodeficient mice and, in immunocompetent settings, mobilizes both exogenous TILs and endogenous CD8+ T cells, enabling tumor eradication and metastasis suppression with 10-fold lower TIL doses than conventional therapies. Collectively, this nanoengineered TIL therapy offers a potential strategy for addressing immune-resistant tumors, showing distinct benefits in stromal-rich settings.
    DOI:  https://doi.org/10.1038/s41467-025-67254-x
  11. Br J Cancer. 2025 Dec 16.
       BACKGROUND: Extranodal extension (ENE) is a recognized adverse prognostic factor in several malignancies, but its pathological basis and clinical significance in lung adenocarcinoma (LUAD) remain poorly understood.
    METHODS: We retrospectively analyzed 105 patients with N1/N2 LUAD who underwent anatomical resection between 2016 and 2020. AI-assisted image analysis quantified tumor-infiltrating lymphocytes (CD8 + , FoxP3 + ), tumor-associated macrophages (CD204 + ), and cancer-associated fibroblasts (CAFs; podoplanin + ) and measured areas of cancer cells and fibrous stroma. In vitro, the effect of CAF quantity on A549 cell invasion was evaluated. Prognostic relevance of ENE was assessed using multivariate and cumulative incidence analyses.
    RESULTS: While primary tumors showed no significant differences in immune or stromal composition, ENE-positive metastatic nodes exhibited a markedly larger fibrous stroma area than ENE-negative nodes (14.2 vs. 4.0 mm², p < 0.001). In vitro, the invasion distance of A549 cells increased when cocultured with higher numbers of CAFs. Clinically, ENE independently predicted poorer overall survival (p = 0.04) and was associated with a higher incidence of distant metastasis in both multivariate (p = 0.03) and cumulative incidence analyses (p = 0.02).
    CONCLUSIONS: ENE represents a pathological prognostic factor characterized by abundant fibrous stroma. It independently predicts distant metastasis and may warrant consideration as a qualitative parameter in N classification for lung adenocarcinoma.
    DOI:  https://doi.org/10.1038/s41416-025-03272-2
  12. Cancer Immunol Immunother. 2025 Dec 18. 75(1): 5
      PIK3CA is the third most common mutated gene in epithelial cancers, behind only TP53 and KRAS. Somatic PIK3CA mutations occur predominantly in the protein's hotspot locations H1047, E545, E542 and N345, and evidence of those alterations eliciting immune responses has been limited. HLA class I-restricted TCRs against E545K and H1047L have, only recently, been identified following in vitro sensitization (IVS) with peripheral blood lymphocytes (PBL) from normal donors. In this study, we examined the immunogenicity of PIK3CA in patients with epithelial cancers of diverse histology, by testing TIL and antigen-experienced PBL against autologous hotspot PIK3CA mutations. Two distinct TCRs specific to PIK3CAN345K, restricted by HLA-class II pair DPB1*04:01/DPA1*01:03, were identified in a patient with colon cancer, following independently a TIL screen and an IVS of memory CD4 + T cells from the peripheral blood. Patient PBL engineered to express each TCR suppressed the in vitro growth of two cell lines modified to express the DPB1*04:01/DPA1*01:03 pair and full length PIK3CAN345K protein. A TCR specific to the PIK3CAE545K, restricted by HLA-DRB1*04:01, was also identified following IVS of memory CD4 + PBL from another patient, with rectal cancer. Autologous PBL, gene engineered to express the PIK3CAE545K-specific TCR, suppressed the in vitro growth of two cancer cell lines, which endogenously expressed the PIK3CAE545K neoantigen. This study identified three PIK3CA-specific TCRs against two shared mutations, restricted by common HLA-class II molecules, using antigen-experienced TIL and memory PBL from patients with epithelial cancers and may further allow the development of off-the-shelf T cell immunotherapy strategies targeting PIK3CA.
    Keywords:  PIK3CA; T cell; TCR; immunotherapy
    DOI:  https://doi.org/10.1007/s00262-025-04156-3
  13. Cancer Invest. 2025 Dec 17. 1-16
      Trophoblast Cell-surface antigen 2 (TROP2), a calcium signal transducer & a transmembrane glycoprotein expression was studied in Triple-Negative Breast Cancer (TNBC) patients. mRNA expression, immunohistochemical and immunofluorescence analysis illustrated a higher TROP2 expression in the TNBC tissues. Assessment of tumor-infiltrating lymphocytes in TNBC samples was done and in silico studies depicted a significant association between TROP2 expression and infiltration of CD4+ cells. Kaplan-Meier survival analysis observed TROP2 expression to be an independent determinant of survival, with better survival benefit in the TROP2 high group. Our study paves the way for newer therapies targeting TROP2 for TNBC patients.
    Keywords:  Indian cohort; Overall survival; TNBC; TROP2; Tumor infiltrating lymphocytes (TILs)
    DOI:  https://doi.org/10.1080/07357907.2025.2595303
  14. Cancer Discov. 2025 Dec 12. OF1
      Early clinical results show that inside-the-body T-cell engineering can trigger rapid chimeric antigen receptor expression and induce deep responses in patients with relapsed multiple myeloma-without the manufacturing required for conventional adoptive cell therapy. Larger studies are needed, however, to determine how off-the-shelf in vivo approaches compare with existing autologous therapies.
    DOI:  https://doi.org/10.1158/2159-8290.CD-NW2025-0112
  15. bioRxiv. 2025 Dec 01. pii: 2025.11.26.690770. [Epub ahead of print]
      Programmed cell death protein 1 (PD-1) is expressed by T cells during progressive multifocal leukoencephalopathy (PML), a life-threatening brain disease caused by the human-only JC polyomavirus. Why PD-1 blockade finds variable success in PML patients is unclear. Brain CD4 + and CD8 + T cells are PD-1 high during mouse polyomavirus (MuPyV) encephalitis. Here, we show that PD-1 loss during MuPyV infection acts in a brain-autonomous manner to increase the magnitude of brain-infiltrating CD4 + and CD8 + T cells and the function of virus-specific CD8 + T cells; in concert, brain virus levels decline and neuroinflammation increases. Deletion of PD-1 in CD4 + T cells, but not CD8 + T cells, recapitulates effects of global PD-1 loss. Single-cell RNA sequencing shows that PD-1-deficient CD8 + T cells cluster as effectors while transcripts associated with proliferation and function are upregulated with loss of PD-1. Thus, CD4 + T cell-intrinsic PD-1 signaling balances antiviral defense against neural injury during polyomavirus CNS infection.
    DOI:  https://doi.org/10.1101/2025.11.26.690770
  16. Cell. 2025 Dec 15. pii: S0092-8674(25)01316-9. [Epub ahead of print]
      While melanoma cells often express a high burden of mutated proteins, the infiltration of reactive T cells rarely results in tumor-eradicating immunity. We discovered that large extracellular vesicles, known as melanosomes, secreted by melanoma cells are decorated with major histocompatibility complex (MHC) molecules that stimulate CD8+ T cells through their T cell receptor (TCR), causing T cell dysfunction and apoptosis. Immunopeptidomic and T cell receptor sequencing (TCR-seq) analyses revealed that these melanosomes carry MHC-bound tumor-associated antigens with higher affinity and immunogenicity, which compete with their tumor cell of origin for direct TCR-MHC interactions. Analysis of biopsies from melanoma patients confirmed that melanosomes trap infiltrating lymphocytes, induce partial activation, and decrease CD8+ T cell cytotoxicity. Inhibition of melanosome secretion in vivo significantly reduced tumor immune evasion. These findings suggest that MHC export protects melanoma from the cytotoxic effects of T cells. Our study highlights a novel immune evasion mechanism and proposes a therapeutic avenue to enhance tumor immunity.
    Keywords:  CD8 T cells; HLA-peptidomics; MHC; TCR-seq; extracellular vesicles; immunotherapy; melanoma; melanosomes; tumor microenvironment; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1016/j.cell.2025.11.020
  17. J Family Med Prim Care. 2025 Nov;14(11): 4631-4634
       Background: Breast cancer is the most common cancer in women across the world. Immune cells can detect and destroy cancer cells. However, cancer cells acquire certain immune evasion mechanisms such as PD-1+ T-cells, to combat immune response. Limited number of studies have shown PD-1+ T-cells as a prognostic marker in breast cancer, but studies in India are far and few.
    Aims: To compare the positivity of PD-1-positive T-cells with known prognostic indicators in breast cancer.
    Materials and Methods: This descriptive, cross-sectional, prospo-retrospective study was conducted at the Department of Pathology of a Tertiary Care Hospital in Uttarakhand, India. A total of 50 cases were taken. Clinicopathological details were noted. On immunohistochemistry, PD-1 was membranous. The distribution pattern and percentage positivity of PD-1-positive T-cells were noted. Pearson Chi-square/Fisher's exact test was used.
    Results: The positivity of PD-1+ T-cells was found to be higher in tumors with luminal B subtype (P = 0.04). Out of the total CD3+ T-cells, 62% were found to be PD-1 positive.
    Conclusion: PD-1 positivity was associated with tumor molecular subtypes. No significant association of PD-1+ T-cells with other known prognostic markers of breast cancer was found.
    Keywords:  Breast carcinoma; programmed death-1; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.4103/jfmpc.jfmpc_320_25
  18. Cell Rep Med. 2025 Dec 16. pii: S2666-3791(25)00588-9. [Epub ahead of print] 102515
      Immune checkpoint blockade (ICB) therapy exerts anti-tumor efficacy mainly by activating intratumoral CD8+ T cells but fails to re-activate terminally exhausted CD8+ T cells. Interleukin-10 (IL-10) has been shown to directly expand and activate these cells and to exert a synergistic effect when combined with ICB. Nevertheless, the clinical application of IL-10 for cancer immunotherapy is restricted by severe hematological toxicity. Here, we design FP008 (anti-PD-1×IL-10M), a clinical-stage fusion protein composed of an anti-PD-1 antibody and an attenuated IL-10 monomer (IL-10M). Mechanistically, the activity and toxicity of IL-10M are significantly reduced, while its therapeutic benefits are enhanced through anti-PD-1-targeted enrichment and cis-activation. Anti-PD-1×IL-10M therapy displays robust anti-tumor activity in various mouse models, including those resistant to anti-PD-1 therapy, and exhibits promising safety in GLP toxicology studies in cynomolgus monkeys. Altogether, reinvigorating exhausted CD8+ T cells in the tumor microenvironment through anti-PD-1×IL-10M represents a promising therapeutic strategy for overcoming anti-PD-1/L1-refractory solid tumors.
    Keywords:  ICB; IL-10 monomer; anti-PD-1; anti-tumor activity; cis-activation; fusion protein; hematological toxicity; terminally exhausted CD8⁺ T
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102515