bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–06–08
fifteen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Front Immunol. 2025 ;16 1601773
      Tumor-infiltrating lymphocytes (TILs) are a diverse population of immune cells that play a central role in tumor immunity and have emerged as critical mediators in cancer immunotherapy. This review explores the phenotypic and functional diversity of TILs-including CD8+ cytotoxic T cells, CD4+ helper T cells, regulatory T cells, B cells, and natural killer (NK) cells-and their dynamic interactions within the tumor microenvironment (TME). While TILs can drive tumor regression, their activity is often hindered by immune checkpoint signaling, metabolic exhaustion, and stromal exclusion. We highlight TIL recruitment, activation, and polarization mechanisms, focusing on chemokine gradients, endothelial adhesion molecules, and dendritic cell-mediated priming. Special emphasis is placed on preclinical models that evaluate TIL function, including 3D tumor spheroids, organoid co-cultures, syngeneic mouse models, and humanized systems. These provide valuable platforms for optimizing TIL-based therapies. Furthermore, we examine the prognostic and predictive value of TILs across cancer types, their role in adoptive cell therapy, and the challenges of translating preclinical success into clinical efficacy. Emerging technologies such as single-cell sequencing, neoantigen prediction, and biomaterial platforms are transforming our understanding of TIL biology and enhancing their therapeutic potential. Innovative strategies-ranging from genetic engineering and combination therapies to targeted modulation of the TME-are being developed to overcome resistance mechanisms and improve TIL persistence, infiltration, and cytotoxicity. This review integrates current advances in TIL research and therapy, offering a comprehensive foundation for future clinical translation. TILs hold significant promise as both biomarkers and therapeutic agents, and with continued innovation, they are poised to become a cornerstone of personalized cancer immunotherapy.
    Keywords:  adoptive cell transfer; experimental models; immunotherapy; tumor microenvironment; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.3389/fimmu.2025.1601773
  2. Front Immunol. 2025 ;16 1524842
      Cervical cancer is a common malignant tumor in women, and human papillomavirus (HPV) infection is a major cause of cervical cancer. Tumor-infiltrating lymphocytes (TILs) are a heterogeneous group of lymphocytes primarily composed of T lymphocytes found within the tumor parenchyma and stroma. These cells can be isolated from tumor tissue, activated, expanded in vitro, and reinfused into the patient to exert an anti-tumor immune effect. As a form of personalized immunotherapy, TILs therapy has shown satisfactory efficacy and safety in advanced recurrent and metastatic cervical cancer, offering new hope to patients with advanced cervical cancer. However, TILs therapy for advanced cervical cancer still faces several limitations and challenges. This article reviews the process and latest developments in TILs therapy for advanced cervical cancer and discusses the challenges in the usage and prospects for this treatment.
    Keywords:  TILs therapy; cervical cancer; human papillomavirus; immunotherapy; tumor-infiltrating lymphocytes (TILs)
    DOI:  https://doi.org/10.3389/fimmu.2025.1524842
  3. bioRxiv. 2025 May 15. pii: 2025.05.11.653315. [Epub ahead of print]
      Neoantigen-reactive peripheral blood lymphocytes (NeoPBL) are tumor-specific T cells found at ultra-low frequencies in the blood. Unlike tumor-infiltrating lymphocytes (TIL), NeoPBL exist in a favorable less dysfunctional phenotypic state in vivo , but their rarity has precluded their effective use as cell therapy. Leveraging a priori knowledge of bona fide neoantigens, we combined high-intensity neoantigen stimulation with bead extraction of neoantigen peptide-pulsed target cells to enable the enrichment of NeoPBL to frequencies comparable to ex vivo cultured TIL over a 28-day period. Throughout this process, NeoPBL demonstrate specific reactivity against autologous tumor organoids and maintain memory-like features, including elevated expression of CD28 and TCF7 . We additionally demonstrate that NeoPBL reactivity is polyclonal, encompassing multiple clonotypes that are detectable within in vivo TIL populations, underscoring physiological specificity for the targeted neoantigens. This streamlined process yields clinically relevant cell doses and enables identification and expansion of blood-derived neoantigen-specific TCRs. By potentially avoiding additional surgical risks and protracted delays of TIL and individualized TCR-engineered methods, the NeoPBL platform may have clinical and practical advantages. Ultimately, NeoPBL combines intrinsic cell fitness, minimal invasiveness and rapidity to potentially facilitate personalized adoptive cell therapy for cancer.
    DOI:  https://doi.org/10.1101/2025.05.11.653315
  4. Front Immunol. 2025 ;16 1507870
      Sarcomas are heterogeneous mesenchymal malignancies classified as soft-tissue sarcomas (STS) and bone sarcomas. Advanced cases respond poorly to standard therapies, highlighting the need for novel strategies. Immunotherapies, including PD-1/PD-L1 inhibitors, adoptive cellular therapies, vaccines, and oncolytic viruses, have shown promise in specific sarcoma subtypes. This review explores these approaches, emphasizing the prognostic significance of immune cells within the tumor microenvironment (TME), such as tumor-associated macrophages (TAMs) and tumor-infiltrating lymphocytes (TILs), and their correlation with clinical outcomes. We also discuss challenges in immunotherapy efficacy, the importance of biomarker-driven personalized therapies, and the potential of a combination regimen with chemotherapy, radiation, and cytokine agents. Overall, this review highlights the evolving role of immunotherapy in advanced sarcomas, the critical influence of the TME, and the need to optimize synergistic treatment approaches to enhance patient outcomes.
    Keywords:  adoptive T cell therapy; immune checkpoint inhibitors; immunotherapy; sarcoma; vaccine
    DOI:  https://doi.org/10.3389/fimmu.2025.1507870
  5. J Clin Oncol. 2025 Jun 02. 101200JCO2500765
    C-144-01 Investigators
      Patients with advanced melanoma resistant to immune checkpoint or BRAF/MEK inhibitors have treatment options with relatively low efficacy. Lifileucel, a one-time autologous tumor-infiltrating lymphocyte cell therapy, was approved in the US based on the pivotal C-144-01 study. A 5-year follow-up of the C-144-01 trial assessed the long-term efficacy and safety of lifileucel. At the cutoff date (November 20, 2024), the ORR was 31.4% (complete response [CR], 5.9%; partial response [PR], 25.5%). Overall, 79.3% of patients had tumor burden reduction; 16 had deepened responses with 4 converting from PR to CR > 1 year after lifileucel infusion; 31.3% of responders completed the 5-year assessment with ongoing responses. The median duration of response was 36.5 months. Responders (n = 48) had lower tumor burden and fewer liver or brain metastases than the overall population. Median overall survival (OS) was 13.9 months, with 5-year OS of 19.7%. Adverse events were consistent with nonmyeloablative lymphodepletion and interleukin-2 safety profiles and declined rapidly within 2 weeks after lifileucel infusion. Most grade 3/4 cytopenias resolved to grade ≤ 2 by day 30. This 5-year analysis demonstrated long-term benefit and meaningful OS with one-time lifileucel therapy, with no additional long-term safety concerns.
    Keywords:  autologous transplantation; skin neoplasms; tumor infiltrating lymphocytes
    DOI:  https://doi.org/10.1200/JCO-25-00765
  6. Br J Surg. 2025 May 31. pii: znaf090. [Epub ahead of print]112(6):
       BACKGROUND: The aim of this study was to describe the impact of surgical resections on tumour-infiltrating lymphocyte (TIL) therapy, based on results from a randomized phase III trial comparing TIL therapy with standard ipilimumab in patients with metastatic melanoma (NCT02278887).
    METHODS: Surgical details of all operations performed in the trial were collected. Location, total number, and size of resected metastases were assessed in relation to successful TIL investigational medicinal product (IMP) manufacture and response to TIL therapy.
    RESULTS: A total of 93 operations were needed to treat 80 patients with TIL therapy. Surgical complications were detected in 17% of operations. These were mostly mild Clavien-Dindo grade I/II complications and one Clavien-Dindo grade IIIa complication. The size or number of resected lesions did not significantly impact the TIL-IMP manufacture failure or response rate. The failure rate of TIL-IMP manufacture from lymph node metastases was 2.8% (1 of 36), which was lower than from subcutaneous metastases (19.4% (6 of 31), P = 0.037) and other sites (15.0% (3 of 20), P = 0.038). Response rates per resected lesion type were 52.9% (18 of 34) for lymph nodes, 40.9% (9 of 22) for subcutaneous lesions, 58.8% (10 of 17) for other lesions, and 60.0% (3 of 5) for combinations of lesions, without statistically significant differences.
    CONCLUSION: Tumour resections for TIL-IMP manufacture lead to limited surgical complications. Manufacture with a therapeutic response was successful using lesions of varying sizes from different anatomical locations, with highest rates for lymph nodes.
    DOI:  https://doi.org/10.1093/bjs/znaf090
  7. Appl Immunohistochem Mol Morphol. 2025 Jun 02.
      In the context of nonsmall cell lung cancer (NSCLC), the immune evasion strategy used by tumor cells involves the expression of immune checkpoint proteins, such as PD-L1, which suppress antitumor T-cell responses. The use of immune checkpoint inhibitors (ICIs) has significantly improved overall survival, overall response rate, and progression-free survival in NSCLC patients. This study aimed to evaluate the concordance of PD-L1 expression in NSCLC patients using tissue microarrays (TMA) as proxies for small biopsies. The degree of concordance among tissue cores and between the cores and the whole slide was reported. Furthermore, the presence of tumor-associated macrophages (TAMs) and tumor-infiltrating lymphocytes (TILs) was analyzed to investigate the correlation between PD-L1 expression and immune cell infiltration. The study included 13 paraffin-embedded tissue samples from patients incidentally diagnosed with lung cancer during COVID imaging studies. Tissue microarrays were constructed using a manual tissue arrayer, and 4 cores of 2 mm diameter of representative areas were selected from the hematoxylin-eosin-stained sections from lung tumor specimens. Immunohistochemical analyses were performed to assess PD-L1 positivity, tumor macrophage infiltrate, and T-cell infiltrate. The density of CD8+ T cells was evaluated as the overall percentage of the area within the borders of the tumors covered by positive immune cells. Results demonstrated that PD-L1 expression showed a high degree of concordance between TMA cores and whole tumor sections, suggesting that small samples could reliably represent whole tumor PD-L1 status. However, the densities of CD8+ T cells and CD68+ macrophages varied significantly. TMA cores typically underrepresented the density of these immune cells compared with whole sections, particularly for CD68+ macrophages, which exhibited lower densities in TMAs, used as proxies for small biopsies. This study contributes to the understanding of how the heterogeneity of PD-L1 expression and immune cell distribution can influence the detection and scoring of these parameters, highlighting the importance of comprehensive immune profiling in guiding personalized cancer immunotherapy.
    Keywords:  COVID; PD-L1; immunotherapy; lung cancer screening; tumor infiltrating lymphocytes
    DOI:  https://doi.org/10.1097/PAI.0000000000001266
  8. Clin Cancer Res. 2025 Jun 03.
       PURPOSE: Clinical trials have shown limited efficacy of anti-programmed cell death protein 1 (PD-1) treatment for glioblastoma (GBM). In this study, we examined the expression of TGFbRI in GBM-infiltrating CD8+ T cells and the characteristics of TGFbRI+CD8+ T cells. We examined the ex vivo effects of co-blockade of PD-1 and TGFb on the functions of GBM-infiltrating CD8+ T cells.
    EXPERIMENTAL DESIGN: Using flow cytometry, we examined the phenotypes of tumor-infiltrating CD8+ T cells from newly diagnosed GBM patients. We performed single-cell RNA/TCR-sequencing to characterize the tumor-infiltrating TGFbRI+CD8+ T cells. We also examined the effects of co-blockade of PD-1 and TGFb on the functions of tumor-infiltrating CD8+ T cells in ex vivo assays.
    RESULTS: GBM-infiltrating CD8+ T cells expressed significantly increased levels of TGFβRI compared to peripheral blood CD8+ T cells. Among tumor-infiltrating CD8+ T cells, TGFbRI+CD8+ T cells exhibited increased expression of immune checkpoint inhibitory receptors, and tumor antigen-specific cells were enriched in TGFbRI+CD8+ T cells. Single-cell profiling revealed that tumor-infiltrating TGFBR1+CD8+ T cells demonstrated more clonal expansion and upregulation of TCR signaling genes compared to TGFBR1-CD8+ T cells. In vitro, anti-CD3 stimulation upregulated TGFbRI expression on CD8+ T cells. GBM patients with a high frequency of TGFbRI+CD8+ T cells presented with increased TGFb signaling intensity. Importantly, combined blockade of PD-1 and TGFb significantly enhanced the functions of tumor-infiltrating CD8+ T cells ex vivo.
    CONCLUSIONS: Our findings provide a basis for further investigation of co-blockade of PD-1 and TGFβ for the treatment of patients with GBM.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-24-2184
  9. J Nanobiotechnology. 2025 Jun 03. 23(1): 413
      Although the programmed cell death protein 1 (PD-1) blockade has been authorized for the treatment of recurrent and metastatic cervical cancer (CC) patients, a significant proportion of CC patients show low objective response rates (ORR) to immune checkpoint blockades (ICBs). Therefore, identifying novel combination treatment strategies to enhance ICBs therapeutic efficacy for CC patients is urgently needed. Here, we discovered that CD39 was highly expressed in exhausted CD8 + T cells from 10 CC patients in our center via single-cell RNA sequencing (scRNA-seq). Furthermore, we validated that CC patients with CD39 highly expressed in CD8 + T cells associated with poor prognosis and immunoevasive subtype of CC both in cohort from our center and the Cancer Genome Atlas (TCGA) database. Moreover, it was also confirmed that CD39-inhibiting not only enhanced the cytotoxicity of CD8 + tumor-infiltrating lymphocytes (TILs) but also promoted the infiltration of B cells through increasing CXCL13 secretion both in vitro experiments and subcutaneous tumor models, thereby amplifying anti-tumor immunity of PD-1 blockade. What was more, we have developed a liposome containing POM-1, which effectively enhanced the anti-tumor effect of POM-1. Our findings provide compelling evidence that targeting CD39 represents a promising "two birds with one stone" strategy for cervical cancer treatment.
    Keywords:  B cells; Cervical cancer (CC); Exhausted T cells; Immune checkpoint blockage (ICB); Tumor microenvironment; αCD39
    DOI:  https://doi.org/10.1186/s12951-025-03500-0
  10. Cell Rep Med. 2025 Jun 02. pii: S2666-3791(25)00214-9. [Epub ahead of print] 102141
      The lack of T cells in tumors is a major hurdle to successful immune checkpoint therapy (ICT). Therefore, therapeutic strategies promoting T cell recruitment into tumors are warranted to improve the treatment efficacy. Here, we report that Fc-optimized anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies are potent remodelers of tumor vasculature that increase tumor-associated high endothelial venules (TA-HEVs), specialized blood vessels supporting lymphocyte entry into tumors. Mechanistically, this effect is dependent on the Fc domain of anti-CTLA-4 antibodies and CD4+ T cells and involves interferon gamma (IFNγ). Unexpectedly, we find that the human anti-CTLA-4 antibody ipilimumab fails to increase TA-HEVs in a humanized mouse model. However, increasing its Fc effector function rescues the modulation of TA-HEVs, promotes CD4+ and CD8+ T cell infiltration into tumors, and sensitizes recalcitrant tumors to programmed cell death protein 1 (PD-1) blockade. Our findings suggest that Fc-optimized anti-CTLA-4 antibodies could be used to reprogram tumor vasculature in poorly immunogenic cold tumors and improve the efficacy of ICT.
    Keywords:  CTLA-4; Fc receptors; antibody engineering; cancer immunotherapy; high endothelial venule; ipilimumab; lymphocyte trafficking; tumor blood vessels; tumor microenvironment; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102141
  11. Semin Oncol Nurs. 2025 Jun 01. pii: S0749-2081(25)00105-6. [Epub ahead of print] 151912
       OBJECTIVES: Adoptive cell therapy (ACT) is a growing personalized immuno-oncology approach, delivered both in standard of care (SOC) and clinical trial (CT) settings. Understanding patient and informal caregivers (ICs) experiences is crucial to optimizing care. This qualitative systematic review explores the ACT experience across three elements: actors (patients and ICs), settings (CT and SOC), and phases of the care continuum.
    METHODS: A systematic search was conducted across Medline, Embase, CINAHL, APA PsycInfo, Cochrane, Web of Science, ProQuest Dissertations & Theses, and Google Scholar up to May 8, 2024. Studies were appraised using the JBI Critical Appraisal Checklist for Qualitative Research, with data extracted and synthesized using a meta-aggregation approach. MAXQDA was used to generate co-occurrence networks between key elements and inductively derived codes. A comparative sentiment analysis highlighted emotional differences between CT and SOC settings.
    RESULTS: Nineteen qualitative studies were included, capturing experiences of patients (n = 19) and ICs (n = 7) receiving chimeric antigen receptor T cell (n = 17) and tumor-infiltrating lymphocyte (n = 2) therapy in CT (n = 13) and SOC (n = 9) settings. Findings revealed phase-specific challenges across physical, cognitive, psychological, emotional, social, financial, professional, communication, and informational domains. These challenges originate from ACT-related toxicities, care pathway complexity, and the novel nature of the therapy.
    CONCLUSIONS: This review identifies the key challenges faced by patients and ICs throughout the ACT care pathway, emphasizing the need for tailored interventions based on the phase and setting, as well as improved support systems.
    IMPLICATIONS FOR NURSING PRACTICES: Recommended strategies include developing decision support tools, establishing caregiver support programs, and implementing navigation services to enhance patient and ICs experiences.
    Keywords:  Adoptive cell therapy (ACT); Caregiver burden; Oncology nursing; Person-centered care; Qualitative studies; Systematic review
    DOI:  https://doi.org/10.1016/j.soncn.2025.151912
  12. J Transl Med. 2025 Jun 03. 23(1): 620
       BACKGROUND: Adoptive transfer of regulatory T cells (Tregs) has provided promising results in treating autoimmune disorders, transplant rejection and graft versus-host disease in early clinical trials. However, major challenges remain for developing a standardized and robust good manufacturing practice (GMP)-compliant cell product which is severely hampered by low frequency of Tregs in circulation and laborious ex vivo expansion.
    METHODS: Paediatric thymuses routinely obtained during heart surgery have been shown by us and others to be a valuable source of large numbers of pure Tregs (Thy-Tregs). Here we show results from our process development approach including systematic laboratory-scale testing of activation reagents, restimulation timing, and cryopreservation to translate our expansion protocol of Thy-Tregs into a clinical grade cell product.
    RESULTS: Thy-Tregs obtained through CD8+ cell depletion and subsequent CD25+ enrichment were expanded with αCD3/αCD28 beads in the presence of Rapamycin and IL-2 for 10-23 days using G-Rex bioreactors. We successfully embedded bead removal and final formulation of a cryopreserved cell product ready to be used at bedside transfusion.
    CONCLUSION: This process has proved the capability of efficiently producing high number of functional Thy-Tregs, which will be administered as cell therapy in children undergoing heart transplantation (ATT-Heart, ISRCTN15374803), and enhancing the potential of using expanded Thy-Tregs for broad-ranging therapeutic applications.
    Keywords:  Clinical trial; GMP; Immuno-therapy; Thy-Tregs
    DOI:  https://doi.org/10.1186/s12967-025-06561-9
  13. Cell Mol Immunol. 2025 Jun 03.
      Upon antigen recognition, CD8+ T cells undergo robust expansion and differentiation to give rise to effector and memory CD8+ T cells. The spatial determinants of the fate of effector and memory CD8+ T cells during acute infection are poorly understood. By integrating single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics, we revealed that naïve CD8+ T cells adopted a divergent trajectory in which they rapidly differentiated into memory precursor (MP) cells and IFN-responsive cells, with the latter representing the entry point of the effector T-cell lineage. In the spleen, monocytes largely colocalized with CD8+ MP cells following antigen stimulation. Specifically, compared with dendritic cells (DCs), the Ly6ChiCCR2+ subset of monocytes promotes memory CD8+ T-cell differentiation. Mechanistically, monocytes express high levels of membrane-bound transforming growth factor-β (TGF-β), which is activated by thrombospondin-1 (TSP-1) to drive the memory CD8+ T-cell program through Smad signaling. Overall, our study reveals a novel spatial mechanism for CD8+ T-cell fate decisions, shedding light on the importance of monocytes in fostering memory CD8+ T-cell development in a cell‒cell contact- and TGF-β-dependent manner.
    Keywords:  Memory CD8+ T cells; Monocytes; Spatial transcriptomics; TGF-β; scRNA-seq
    DOI:  https://doi.org/10.1038/s41423-025-01299-2
  14. Front Immunol. 2025 ;16 1579205
       Introduction: Medullary thyroid carcinoma (MTC) is a rare and aggressive thyroid cancer with a challenging prognosis. While the immune microenvironment plays a crucial role in cancer progression, its role in MTC remains underexplored compared to more common thyroid cancers.
    Methods: this study investigates the immune landscape of MTC by systematically evaluating immune cell infiltration and expression of immune markers across various tissue topographies. We utilized advanced immunohistochemical techniques to analyze tissue samples from 24 MTC patients, focusing on the tumor core, interface with healthy tissue, adjacent normal thyroid tissue, and lymph node metastases.
    Results: our findings reveal a distinct immune profile with increased CD3+, CD4+, CD8+ and CD20+ lymphocytes in normal tissues adjacent to tumors and a notable presence of granzyme B+ cells in the tumor interface, particularly in patients with structural disease. Additionally, we observed a significant enrichment of mast cells in metastatic tissues.
    Discussion: these results highlight the complex and spatially dependent immune landscape of MTC, suggesting implications for targeted immunotherapy. This study provides novel insights into the immune microenvironment of MTC and emphasizes the need for further research to elucidate its impact on disease progression and therapeutic response.
    Keywords:  PD-L1 expression; immune microenvironment; mast cells; medullary thyroid carcinoma (MTC); tumor-infiltrating lymphocytes (TILs)
    DOI:  https://doi.org/10.3389/fimmu.2025.1579205
  15. Clin Transl Immunology. 2025 ;14(6): e70025
       Objectives: Chimeric antigen receptor (CAR) T-cell therapies have revolutionised the treatment of blood-based malignancies. The use of manual CAR T-cell manufacturing methods is one of the challenges that contributes to these delays. As CAR T therapy emerges as a potential first- or second-line treatment option for these cancers, the demand for these therapies continues to rise. However, challenges persist in ensuring that the patients who need these therapies receive them in a timely manner. Automated CAR T-cell manufacturing methods that use software to control the equipment used in the process can help overcome the roadblocks associated with manual manufacturing, ultimately enabling a reduction in variability, increased efficiency, improved product quality and better data management. This paper aims to present an end-to-end semi-automated methodology for manufacturing CAR T cells using the Cell Therapy Systems (CTS™) Cellmation software - an off-the-shelf software solution - to control physically connected modular cell therapy instruments that eliminates the roadblocks associated with manual manufacturing.
    Methods: T cells from healthy donors were isolated and processed into CAR T cells using a semi-automated, connected, multi-instrument setup that leveraged electroporation and a CRISPR/Cas system for delivering the CD19-CAR construct to the T cells. Flow cytometry was used to assess cell type composition, cell viability and expression of T-cell activation markers throughout the process. We also measured exhaustion marker expression on T cells, T-cell receptor (TCR) knock-out, CAR knock-in and cytotoxic activity against NALM6 cells.
    Results: The results demonstrated the successful generation of functional CAR T cells using a semi-automated instrument workflow. The results were similar to the results from CAR T cells manufactured using non-automated processes; however, the successful connection and control of the instruments using automated software present an exciting opportunity for process developers and manufacturers who want to reduce manual touchpoints in their cell therapy manufacturing process.
    Conclusion: The method that we describe in this paper could be beneficial to process development and manufacturing teams that might require flexibility in their CAR T cell manufacturing workflow and want to take advantage of modular systems that can be automated using the Cellmation software to reduce the problems associated with manual handling.
    Keywords:  CAR T cells; CAR T‐cell manufacturing; automation; cell therapy
    DOI:  https://doi.org/10.1002/cti2.70025