bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–07–20
twelve papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Eur J Cancer. 2025 Jul 13. pii: S0959-8049(25)00422-8. [Epub ahead of print]227 115640
       AIMS: Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TILs) has demonstrated benefit for patients with advanced melanoma refractory to first-line immune checkpoint inhibitors (ICIs). However, predictive biomarkers for TIL therapy response are limited. Preclinical studies suggest that emotional distress (ED) may impair antitumor immune responses. We conducted a post-hoc analysis of the phase III TIL trial (NCT02278887) to evaluate the association between pretreatment ED and TIL therapy outcomes.
    METHODS: ED was evaluated using a composite score derived from the Impact of Event Scale and the emotional functioning subscale of the EORTC QLQ-C15-PAL. Patients were stratified into elevated ED (n = 32) or normal ED (n = 40) groups using a median split approach. To evaluate whether the obtained results were dependent on the ED classification method, a sensitivity analysis was conducted to compare TIL therapy outcomes between patients with composite ED scores in the lowest tertile (normal ED, n = 26) and highest tertile (elevated ED, n = 25).
    RESULTS: Elevated ED was associated with a reduced objective response rate (37 % vs. 69 %; adjusted odds ratio, 0.28; 95 % confidence interval [CI], 0.19-0.75; P = 0.013) and decreased progression-free survival rates at 1-year (31 % vs. 58 %) and 2-years (16 % vs. 35 %; adjusted hazard ratio, 1.84; 95 % CI, 1.03-3.26; P = 0.038). Sensitivity analysis using the tertile grouping method showed a similar trend (odds ratio, 0.35; 95 % CI, 0.11-1.08, P = 0.072).
    CONCLUSION: Our findings suggest that elevated ED before TIL therapy is associated with reduced therapeutic outcomes, underscoring the need for further investigation into the stress-immune-cancer axis.
    Keywords:  Clinical outcomes; Emotional distress; Immunotherapy; Melanoma; Survival
    DOI:  https://doi.org/10.1016/j.ejca.2025.115640
  2. Thorac Cancer. 2025 Jul;16(14): e70135
       BACKGROUND: Despite great expectations regarding the use of tumor-infiltrating lymphocytes (TILs) in predicting the effects of immunotherapies and prognosis, knowledge about TILs remains inadequate for clinical application. We re-evaluated the clinicopathological significance of CD8+ tumor-infiltrating lymphocytes (CD8 + TILs) in lung adenocarcinoma from a novel perspective and through a more objective approach using cell counting software.
    METHODS: Among patients with surgical resection of lung adenocarcinoma in 2011-2017, 156 patients with pathological stage IB-III were immunohistochemically studied to evaluate CD8 + TILs in the tumor stroma. The impact of CD8 + TILs on relapse-free survival was analyzed by Kaplan-Meier survival and multivariate Cox proportional hazards analyses.
    RESULTS: Our analysis showed that patients with large numbers of CD8 + TILs in the stroma are not a homogeneous population and include subpopulations with a poor prognosis. Furthermore, our study showed important findings about the overall inflammatory status in the tumor microenvironment based on the association between the number of CD8 + TILs and the frequency of vascular invasion. Additionally, CD8 + TILs were shown to potentially be more effective than PD-L1.
    CONCLUSIONS: Our study demonstrated that in lung adenocarcinoma, even among populations with abundant CD8 + TILs in the tumor stroma, there may be a poor prognostic subgroup. Furthermore, we revealed a partial yet important relationship between CD8 + TILs and the tumor microenvironment. A more detailed investigation into the significance of CD8 + TILs may lead to a deeper understanding of the inflammatory status of the tumor microenvironment and ultimately contribute to the identification of appropriate biomarkers for prognostic prediction and assessing the efficacy of immune checkpoint inhibitors.
    Keywords:  biomarker; immune checkpoint inhibitors; lung cancer; tumor microenvironment; tumor‐infiltrating lymphocytes
    DOI:  https://doi.org/10.1111/1759-7714.70135
  3. J Vis Exp. 2025 Jun 27.
      Colorectal cancer (CRC) is the third most common cancer worldwide. Tumor-infiltrating lymphocytes (TILs) have been identified as an important prognostic marker in CRC. The therapeutic application of TILs has already shown promising results in melanoma and cervical cancer. However, their use in CRC therapy remains in an exploratory phase. A suitable in vitro model to evaluate TIL efficacy is currently unavailable, hindering further advancements in this field. Patient-derived organoid (PDO) models, which closely retain the characteristics of the original tumor tissue and reflect inter-patient heterogeneity, provide an excellent platform for studying the interaction between CRC and TILs. In this study, a method is described to establish a patient-derived CRC organoid model from freshly resected tumor tissue, followed by isolation and expansion of TILs. This system allows co-culture of CRC organoids and TILs, enabling the assessment of TIL-mediated cytotoxicity and immune responses. By analyzing TIL killing efficacy on organoids, the potential outcomes of TIL-based immunotherapy for personalized CRC treatment can be predicted. Moreover, further engineering of TILs may enhance their anti-tumor efficacy, offering a promising strategy for developing more effective cellular therapies. This PDO-TIL co-culture model provides a powerful tool for preclinical evaluation of TIL therapies and personalized treatment strategies in CRC.
    DOI:  https://doi.org/10.3791/68346
  4. Oncoimmunology. 2025 Dec;14(1): 2532662
      Cancer immunotherapy predominantly targets CD8 T cells, but recent evidence highlights the importance of CD4 T cells in adoptive cell therapy (ACT). The TAM receptor MerTK regulates immune responses and has been shown to provide costimulatory signals in CD8 T cells. However, its role in CD4 T cells remains poorly understood. Here, we demonstrate that ProS1-MerTK signaling is upregulated in activated CD4 T cells, where it enhances central memory formation, metabolic fitness, and proliferation. Mechanistically, ProS1-MerTK signaling was linked to type 1 immune responses, suggesting a regulatory role in CD4 T cell polarization. Using CRISPR-Cas9-mediated knockout, we found that loss of MerTK reduced CD4 T cell fitness, function, and polarization. Furthermore, when ProS1 was added during the expansion of tumor-infiltrating lymphocytes (TILs) from advanced melanoma biopsies, it showed potential to promote favorable CD4 T cell memory and helper phenotypes, increase stemness, and reduce exhaustion - features associated with improved responses to ACT. These findings establish ProS1-MerTK as a key pathway for modulating CD4 T cell functionality and highlight its therapeutic potential to enhance TIL-based ACT outcomes.
    Keywords:  Adoptive cell therapy; T cell; TAM-receptor; immunotherapy; melanoma; tumour infiltrating lymphocyte
    DOI:  https://doi.org/10.1080/2162402X.2025.2532662
  5. J Immunother Cancer. 2025 Jul 15. pii: e011657. [Epub ahead of print]13(7):
       BACKGROUND: Tumor-derived proprotein convertase subtilisin/kexin type 9 (PCSK9) facilitates tumor progression, but the role of immune cell-intrinsic PCSK9 in tumor control remains unclear.
    METHODS: Orthotopic models of pancreatic cancer and melanoma in Pcsk9-deficient mice were established and tumor-infiltrating immune cells were analyzed using single-cell RNA sequencing and flow cytometry. The effect of genetic disruptions of PCSK9 on murine CD8+ T cells and human chimeric antigen receptor (CAR)-T cells was evaluated both in vitro and in vivo.
    RESULTS: Ablation of host Pcsk9 remarkably suppressed tumor growth and prolonged the survival of tumor-bearing mice, while tumor cells still express PCSK9. The enhanced tumor suppression in Pcsk9-deficient mice depended on CD8+ T cells. Notably, PCSK9 expression was induced in CD8+ tumor-infiltrating lymphocytes (TILs). Consequently, Pcsk9 ablation potentiated the antitumor capacity of CD8+ T cells, showing increased intratumoral infiltration and improved cytotoxic function, along with higher proportions of both effector-memory precursor exhausted (TPEX) and terminally exhausted (TTEX) CD8+ TILs. Additionally, disruption of PCSK9 in both murine CD8+ T cells and human CAR-T cells, synergistic with PD-1 blockade, promoted tumor suppression.
    CONCLUSION: These findings indicate that PCSK9 inhibits the antitumor function of CD8+ T cells, suggesting it may be a promising target for enhancing T-cell-based cancer immunotherapy.
    Keywords:  Adoptive cell therapy - ACT; Immunotherapy; T cell
    DOI:  https://doi.org/10.1136/jitc-2025-011657
  6. Clin Exp Med. 2025 Jul 18. 25(1): 254
      Tumor-infiltrating lymphocyte (TIL) therapy represents an emerging and promising approach in the field of cancer immunotherapy, harnessing the body's own immune cells to target and eliminate cancer cells. This review provides an in-depth analysis of TIL therapy, focusing on its mechanisms, clinical applications, challenges, and future prospects. TILs, particularly CD8 + T cells, are isolated from a patient's tumor, expanded ex vivo, and reinfused to promote anti-tumor immunity. The success of TIL therapy has been demonstrated in various cancers, particularly melanoma, where it has led to durable responses and, in some cases, complete remission. However, significant challenges remain, including the heterogeneity of TIL populations, difficulties in large-scale expansion, and immune-related adverse events. We also explore current strategies aimed at overcoming these limitations, including genetic modification of TILs, combinatory approaches with checkpoint inhibitors, and optimization of tumor infiltration protocols. With ongoing research and technological advancements, TIL therapy holds substantial promise as a cornerstone of personalized cancer treatment. This review synthesizes the current landscape of TIL therapy, providing insights into its clinical efficacy, potential for broader application, and the future of cancer immunotherapy.
    Keywords:  Antigens; Immunotherapy; Lymphocytes; Melanoma; TIL therapy; Tumor
    DOI:  https://doi.org/10.1007/s10238-025-01574-6
  7. Cancer Med. 2025 Jul;14(14): e71054
       BACKGROUND: Tumor-infiltrating lymphocytes (TILs) have been proven to be important prognostic factors for various tumors. However, their prognostic significance within the context of esophageal squamous cell carcinoma (ESCC) remains inadequately explored. This study aims to assess the prognostic potential of TILs in ESCC using deep learning (DL) methods.
    MATERIALS AND METHODS: We retrospectively enrolled 626 pathologically confirmed ESCC patients from two research centers. Their digital whole-slide imaging (WSI) and corresponding clinical information were collected. Subsequently, the DL method was employed to identify the tumor margin and TILs within the WSI. Tissue was divided into intratumor, peritumoral, and stromal regions based on their distance from the tumor margin. TILs were counted in each region. Optimal cut-off values of TILs were determined using the X-tile software. To mitigate selection bias and intergroup heterogeneity, a propensity score matching (PSM) analysis was employed. Survival analysis was performed using Kaplan-Meier curves and the log-rank test. The Cox proportional hazards regression model was used to identify independent prognostic factors.
    RESULTS: We classified patients based on the cell counts and cut-off values of intratumor-infiltrating lymphocytes (I-TILs) and peritumoral infiltrating lymphocytes (P-TILs). Patients with high I-TILs and P-TILs were defined as those whose counts of both I-TILs and P-TILs exceeded the determined cutoff value. Patients with high I-TILs and P-TILs showed significantly better overall survival (OS, p = 0.0092) and recurrence-free survival (RFS, p = 0.0088) than patients with low I-TILs and P-TILs after PSM. Multivariable Cox proportional hazards regression further supported this conclusion and recognized I-TILs and P-TILs as independent prognostic factors (p = 0.0136, hazard ratio = 0.63 for OS; p = 0.0098, hazard ratio = 0.63 for RFS).
    CONCLUSION: In the present study, we identified the quantitative distribution of TILs in ESCC patients with the help of the DL method. We established that I-TILs and P-TILs serve as independent prognostic factors for these patients. Further studies should focus on the lymphocyte subgroups and make better use of the spatial information to improve the predictive efficacy of TILs.
    Keywords:  deep learning; esophageal squamous cell carcinoma; prognostic factors; tumor‐infiltrating lymphocytes
    DOI:  https://doi.org/10.1002/cam4.71054
  8. Cytotherapy. 2025 Jun 21. pii: S1465-3249(25)00746-7. [Epub ahead of print]
       INTRODUCTION: The prognosis of stage IV gastrointestinal (GI) carcinomas is poor with a 15% five-year survival rate for colorectal carcinomas. To improve efficacy of tumor infiltrating lymphocytes (TIL), we isolated mutation-reactive autologous TIL and employed CRISPR/Cas9 to knockout (KO) the intracellular checkpoint protein CISH, which has been shown to enhance T cell expansion, functional avidity, and cytokine polyfunctionality, with consequent durable regression of established tumors in an animal model.
    MATERIALS & METHODS: TIL cultures were initiated from resected tumor fragments and maintained for six weeks before harvest and cryopreservation. Candidate neoantigens were nominated by exome sequencing and peptides were used to identify mutation reactive (MR) TIL. Selected MR TIL were thawed and allowed to recover for 24-36 h in media with 10% AB serum, 6000 IU/mL IL-2, and 5 ng/mL IL-7 and IL-15 followed by stimulation with plate-bound anti-CD3/soluble anti-CD28 for 4 days. CISH KO was performed by electroporation of Cas9 mRNA and chemically modified single guide RNA. Between 5 -7.5 million viable cells were added to each 100 cm2 G-Rex vessel containing 600 mL expansion media (with allogeneic feeder MNC:TIL = 100:1) and incubated for 6-8 days. Cultures were evaluated and split according to cell concentration criteria (and dose cohort) and incubated for an additional 6-8 days. On day 14, all of the cells were harvested, washed with buffer and cryopreserved (5% DMSO). Lot release testing included: viability, %CD3+, cytology review, Gram stain, sterility, endotoxin, mycoplasma, and interferon gamma (IFN-γ) production. Additional testing included DNA sequencing to determine genomic CISH editing efficiency and a Western blot for determination of CISH protein loss.
    RESULTS: Patients with GI cancers (colon [10], rectal [8], pancreatic [1], and esophageal [1]) underwent tumor collection. Nineteen of 22 tumor biopsies sampled from 20 patients total proceeded to KO/expansion. Final TIL product results (mean [SD], median [range]) were: viable count (x 1010) -3.25 (3.67), 1.95 (0.018-12.40); viable TIL fold expansion -327.1 (364.8), 153.1 (8-1454); % viability - 76 (13), 78 (43-92); % CD3 -94.4 (5.4), 95.8 (78.6-99.4); % CISH KO efficiency - 75 (29), 87 (0-96); % editing efficiency - 59.9 (24.8), 66.9 (0.4-86). Viability fell below 70% for five TIL products. All other lot release testing has met specification. Thirteen patients have received TIL; six patients were not treated due to disease progression prior to anticipated infusion.
    CONCLUSION: The translation of CRISPR/Cas9-based CISH KO MR TIL from the basic research lab to current good manufacturing practices The (cGMP) facility was successful, allowing for optimized, large-scale expansion in support of a first-in-human clinical trial to treat patients with metastatic GI cancers (ClinicalTrials.gov Identifier: NCT04426669).
    Keywords:  CISH (cytokine-inducible SH2-containing protein); CRISPR/cas9; Tumor-infiltrating lymphocytes (TIL); cGMP manufacturing; gastrointestinal cancer
    DOI:  https://doi.org/10.1016/j.jcyt.2025.06.007
  9. Cancers (Basel). 2025 Jun 23. pii: 2094. [Epub ahead of print]17(13):
      Introduction: The limited value of TNM staging in locally advanced (LA) nasopharyngeal carcinoma (NPC), defined as stage III or IV NPC, highlights the need for prognostic markers. Here, we aimed to evaluate the ability of the cancer stem cell markers, BMI1, ALDH1, CD44, and CD24, and the epithelial-mesenchymal transition marker, vimentin, as novel prognostic markers in LA-NPC. Methods: A cohort of 83 patients with LA-NPC, previously recruited for another trial with a different goal, was used. The marker expression in tissue sections was evaluated using immunohistochemistry, and the association of expression with survival outcomes was analyzed using the Kaplan-Meier method and Cox regression analysis. Results: The expression of BMI1 and ALDH1 was not associated with survival. The tumoral expression of CD24, CD44, and vimentin was significantly associated with disease-free survival (DFS), whereas only the expression of CD24 was also significantly associated with overall survival (OS). Indeed, CD24 expression emerged as an independent prognostic factor as it was associated with survival in univariate and multivariate Cox regression analysis. Interestingly, combining the status of CD24 expression in tumor cells with the density of CD3+ tumor-infiltrating lymphocytes (TIL), a microenvironment/immune-related marker, identified a high-risk subgroup with the worst DFS and OS. Conclusions: These results suggest the utility of combining tumoral CD24 expression and the CD3+ TIL density as a prognostic factor in LA-NPC, with potential application in disease management and future trial design. Our findings also highlight the potential of combining immune- and cancer stem cell-related factors in personalized treatment strategies for cancer.
    Keywords:  CD3; nasopharyngeal carcinoma; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.3390/cancers17132094
  10. Cancers (Basel). 2025 Jul 01. pii: 2210. [Epub ahead of print]17(13):
      Immune checkpoint inhibition is a cornerstone of bladder cancer therapy, but its efficacy in non-urothelial subtypes of bladder cancer is limited, and the prognosis remains poor. Therefore, we investigated the potential of the immune checkpoint molecules TIM-3, TIGIT, and LAG-3 in squamous-cell carcinoma (SCC) and adenocarcinoma (ADENO) of the urinary bladder. Tumor-infiltrating lymphocytes (TILs) showed a high expression of TIM-3 and TIGIT in both SCC and ADENO, while LAG-3-positive TILs were absent in ADENO and present in 46% of SCC. Quantitative analysis revealed age-independent expression of TIM-3 in SCC (r = -0.001, p = 0.997) and ADENO (r = 0.135, p = 0.549), with increasing age correlating with higher expression of TIGIT (r = 0.157, p = 0.242) and LAG-3 (0.106, p = 0.436) in the SCC cohort and of TIGIT (r = 0.276, p = 0.214) in the ADENO cohort. Male patients showed increased TIGIT scores in ADENO (p < 0.01). Of note, a high infiltration of TIM-3-TILs (p = 0.048) correlated with worse progression-free survival in SCC. These results highlight the differential expression of co-inhibitory receptors in non-urothelial bladder cancer subtypes and provide preclinical evidence for new therapeutic targets. Biomarker testing prior to clinical trials is essential for identifying the most suitable patients for targeted immunotherapy.
    Keywords:  LAG-3; TIGIT; TIM-3; adenocarcinoma; bladder cancer; coinhibitory receptor; squamous-cell carcinoma; variant histology
    DOI:  https://doi.org/10.3390/cancers17132210
  11. Cancers (Basel). 2025 Jul 02. pii: 2225. [Epub ahead of print]17(13):
      Cell-based immunotherapy is a promising treatment strategy for cancer. Particularly in the case of solid tumors, however, this strategy only benefits a minority of patients. A critical limitation to immunotherapy is T cell exhaustion, a terminal differentiation state characterized by loss of self-renewal and cytotoxic capacity. For over a decade, regenerative immunology approaches to overcome exhaustion and restore stem-like features of T cells have been pursued. The reprogramming of tumor-specific T cells back to a less-differentiated, stem-like state using induced pluripotent stem cell (iPSC) technology has been viewed as a powerful and highly appealing strategy to overcome the limitations imposed by exhaustion. However, clinical translation of these approaches has been stymied by the requirement for subsequent iPSC-to-T cell re-maturation strategies, vanishingly low efficiencies, and resource-intensive cell culture protocols. In this review, we discuss the emergence of transcription factor reprogramming to iPSCs, contemporary techniques for T cell reprogramming, as well as techniques for re-differentiation into mature T cells. We discuss the potential clinical utility of T cell reprogramming and re-maturation strategies alongside progress and major roadblocks toward clinical translation. If these challenges can be addressed, transcription factor reprogramming of T cells into iPSCs and subsequent re-maturation into tumor-specific stem-like T cells may represent an incredibly efficacious approach to cancer immunotherapy.
    Keywords:  T cell exhaustion; adoptive cellular therapy; immunotherapy; stemness; transcription factor reprogramming
    DOI:  https://doi.org/10.3390/cancers17132225
  12. Immunotargets Ther. 2025 ;14 697-718
       Purpose: Aspartate β-hydroxylase (ASPH) contributes to carcinogenesis by promoting tumor cell proliferation, migration, and invasion. The enzymatic activity of ASPH can be inhibited by small molecule inhibitors that have been shown to have anti-metastatic activity in rodent models. ASPH has also been shown to inhibit the activation of natural killer (NK) cells. Therefore, this study aimed to investigate the effect of ASPH inhibition on the induction of anti-tumor immunity and to analyze the immune cells involved.
    Methods: In the mouse TC-1/A9 model characterized by reversible downregulation of major histocompatibility class I (MHC-I) molecules, ASPH inhibition was combined with stimulation of innate and/or adaptive immunity, and the anti-tumor response was analyzed by evaluation of tumor growth, in vivo depletion of immune cell subpopulations, and ELISPOT assay. Characteristics of immune cells in the spleen and tumor were determined by flow cytometry and single-cell RNA sequencing (scRNA-seq).
    Results: ASPH inhibition did not reduce tumor growth or promote the anti-tumor effect of innate immunity stimulation with the synthetic oligonucleotide ODN1826, but it significantly enhanced tumor growth reduction induced by DNA vaccination. In vivo immune cell depletion suggested that CD8+ T cells played a critical role in this immunity stimulated by combined treatment with ASPH inhibition and DNA vaccination. ASPH inhibition also significantly enhanced the specific response of CD8+ T cells induced by DNA vaccination in splenocytes, as detected by ELISPOT assay, and reduced the number of regulatory T cells in tumors. scRNA-seq confirmed the improved activation of CD8+ T cells in tumor-infiltrating cells after combined therapy with DNA vaccination and ASPH inhibition. It also showed activation of NK cells, macrophages, and dendritic cells in tumors.
    Conclusion: ASPH inhibition stimulated T-cell-mediated adaptive immunity induced by DNA vaccination. Different types of lymphoid and myeloid cells were likely involved in the activated immune response that was efficient against tumors with MHC-I downregulation, which are often resistant to T-cell-based therapies. Due to different types of activated immune cells, ASPH inhibition could improve immunotherapy for tumors with various MHC-I expression levels.
    Keywords:  ASPH; adaptive immunity; cancer immunotherapy; scRNA-seq; tumor microenvironment
    DOI:  https://doi.org/10.2147/ITT.S530987