bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–06–22
seventeen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Med Oncol. 2025 Jun 15. 42(7): 261
      Adoptive cell therapy (ACT) is a ground-breaking development in cancer treatment that uses modified immune cells to target and eradicate tumors precisely. ACT is a type of immunotherapy in which T cells are genetically manipulated to produce chimeric antigen receptor (CAR) T cells, tumor-infiltrating lymphocytes (TILs), and T cell receptors. CAR-T cell therapy, with its promising effects, has transformed the area of ACTs, notably for hematologic malignancies. ACT is not ideal, and it can cause significant side effects, limiting its use in clinical trials. One of the most promising approaches to reducing side effects is to give adoptive T cells the ability to target neoantigens, which are unique to tumor cells. In this review, we focused on the principles, benefits, challenges, and pre-clinical, translational, and clinical research on ACT, as well as safety concerns such as cytokine release syndrome and neurotoxicity. We also discussed combination approaches, personalized approaches, and emerging technologies involved in maximizing ACT efficacy.
    Keywords:  Adoptive cell therapy; CAR-T cells; Clinical studies; Emerging technologies; Translational research; Tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1007/s12032-025-02808-z
  2. Cancer Immunol Immunother. 2025 Jun 19. 74(8): 244
       BACKGROUND: Adoptive cell therapy with tumor infiltrating lymphocytes is FDA approved for metastatic melanoma. TIL from patients with melanoma and factors relating to growth and reactivity have been studied; however, this has not been explored in patients with epithelial cancers.
    PATIENTS AND METHODS: Metastatic epithelial tumors resected for TIL growth from 2014 to 2023 were analyzed. Two hundred and ninety-one operations were performed to collect TIL for potential treatment. Of these, 263 harvests were each processed for up to 24 individual fragment cultures and screened for neoantigen recognition of the expressed products of cancer mutations. Patient and tumor characteristics were collected. Endpoints were growth (defined as more than half of all fragment cultures expanded for viable cryopreservation) and patient-specific neoantigen reactivity (release of interferon-γ in cocultures measured by ELISpot and 4-1BB upregulation on flow cytometry).
    RESULTS: TIL fragments reached adequate growth for screening by 21 days. Metastatic resections from lung were more likely to grow TIL than all other resection sites combined (95%, p = 0.0011), while hepatic resections were less likely to grow (69%, p < 0.0001). One hundred and thirty-five patients (51%) had highly reactive TIL, 68 (26%) had weakly reactive TIL, and 60 (23%) had TIL with no neoantigen reactivity. Patients with prior exposure to immune checkpoint blockade therapy were less likely to have highly reactive TIL (p = 0.0325). Metastatic resection site impacted TIL reactivity against neoantigens, with those harvested from the lung more likely to show any reactivity (83%, p = 0.0180), as well as high reactivity (59%, p = 0.0066).
    CONCLUSIONS: Prior immune checkpoint therapies reduced the likelihood of having highly reactive TIL. Neoantigen reactivity was more common in TIL from thoracic resections versus other sites. Conversely, hepatic lesions yielded TIL less likely to grow and with less reactivity. These results contribute to improved strategies for sequencing TIL with other therapies and planning TIL harvests for patients with epithelial cancers.
    Keywords:  Adoptive cell transfer; Metastatic solid tumors; Tumor infiltrating lymphocytes
    DOI:  https://doi.org/10.1007/s00262-025-04091-3
  3. Front Immunol. 2025 ;16 1601188
       Introduction: T-cell exhaustion is a major mechanism of immune evasion. Recently, the therapeutic and prognostic implications of progenitor exhausted T cells (Tpex) and terminally exhausted T cells (Ttex) have been explored in various cancer types. This study explored the immunogenomic characteristics and prognostic implications of Tpex and Ttex in colorectal cancers (CRCs).
    Methods: We performed multiplex immunofluorescence (mIF) using antibodies against CK, CD3, CD8, TCF1, and FOXP3 to assess diverse subsets of tumor-infiltrating lymphocytes (TILs) in 517 patients with stage III or high-risk stage II CRCs. We compared the infiltration level of these TIL subsets with the genetic profiles of CRCs, including microsatellite instability (MSI), tumor mutational burden (TMB), and mutations in 40 tumor-associated genes across five biological pathways.
    Results: CD8+ T cell density, the CD8/CD3 ratio, and the Ttex/CD8+ T cell ratio were elevated in microsatellite instability-high and tumor mutational burden-high tumors. Survival analysis showed that, higher CD8+ T cell density, higher regulatory T cell/CD3+ T cell ratio, and higher Ttex/CD8+ T cell ratio exhibited better 5-year relapse-free survival (RFS) rates. When tumors were categorized into CD8-high, CD8-low/Ttex-low, and CD8-low/Ttex-high groups, the CD8-high and CD8-low/Ttex-high groups showed better 5-year RFS than the CD8-low/Ttex-low group.
    Discussion: Ttex infiltration is associated with MSI and TMB status and may serve as a prognostic marker of CRCs.
    Keywords:  colorectal cancer; exhausted T cell; microsatellite instability; prognosis; tumor mutational burden
    DOI:  https://doi.org/10.3389/fimmu.2025.1601188
  4. STAR Protoc. 2025 Jun 14. pii: S2666-1667(25)00301-6. [Epub ahead of print]6(3): 103895
      We present a protocol to generate immunocompetent 3D tumor-on-chip models from human solid tumors, enabling more accurate therapy response assessment than traditional 2D assays. We outline the isolation and culture of autologous tumor cells, CD8+ tumor-infiltrating lymphocytes, and cancer-associated fibroblasts, followed by their encapsulation in a 3D biomimetic matrix within microfluidic devices and subsequent video microscopy. The protocol is adaptable to other tumor types, including breast and colon cancer. For complete details on the use and execution of this protocol, please refer to Veith et al.1.
    Keywords:  Cancer; Cell culture; Cell isolation; Immunology; Tissue Engineering
    DOI:  https://doi.org/10.1016/j.xpro.2025.103895
  5. Discov Oncol. 2025 Jun 20. 16(1): 1170
      We aimed to investigate the genomic and immune microenvironmental characteristics and their prognostic value in limited-stage small cell lung cancer (SCLC). Whole exome sequencing and multiplex immunofluorescence analysis were conducted on 38 patients diagnosed with limited-stage SCLC. The two most frequently mutated cancer-related genes observed were RB1 (73.68%) and TP53 (63.16%). However, none of the cancer-related genes, including RB1 and TP53, were associated with prognosis. Furthermore, genomic factors such as tumor mutation burden, copy number instability, and mutant-allele tumor heterogeneity were unrelated to prognosis. Approximately 52.63% (20/38) of cases exhibited PD-L1 expression (combined positive score > 1). The average percentage of CD8-positive tumor-infiltrating lymphocytes (TILs) was 3.27%, with a range spanning from 0.04 to 18.96%. Survival analyses showed that PD-L1 positivity, a high proportion of CD8-positive TILs, and wild-type PI3K pathway were significantly associated with better survival. A predictive prognostic model was further developed based on these three biomarkers, resulting in more accurate stratification of patients according to disease-free survival (DFS, hazard ratio (HR) = 2.020, P < 0.001) and overall survival (OS, HR = 2.344, P < 0.001). Moreover, PD-L1 negative patients who did not undergo adjuvant chemotherapy exhibited significantly improved OS (P = 0.029) and a favorable trend in DFS (P = 0.053) compared to those who underwent adjuvant chemotherapy. In conclusion, this study analyzed the genomic and immune microenvironment characteristics of limited-stage SCLC and constructed a prognostic model based on PD-L1 expression, CD8-positive TILs, and PI3K pathway mutation, which may potentially contribute to the clinical management of limited-stage SCLC. Clinical trial number: Not applicable.
    Keywords:  CD8; Genomic mutation; PD-L1; PI3K pathway; Prognostic biomarker; Small cell lung cancer
    DOI:  https://doi.org/10.1007/s12672-025-02925-1
  6. J Egypt Natl Canc Inst. 2025 Jun 17. 37(1): 51
      Triple-negative breast cancer (TNBC) accounts for 10-20% of all breast cancers. These tumors are heterogeneous, highly aggressive, and associated with a poor prognosis and a high risk of recurrence. In both hematologic and solid malignancies, immune checkpoint inhibitors (ICIs) have demonstrated the ability to enhance long-term survival and sustain robust anti-tumor responses. Immunotherapy has also been introduced as a treatment option for TNBC, a subtype characterized by a high presence of intra-tumoral tumor-infiltrating lymphocytes (TILs) and stromal immune cells. This heightened immune activity within TNBC serves as a prognostic marker, indicating a potential for better responses to immunotherapy due to increased tumor immune infiltration. This review provides an overview of the current landscape of immunotherapy in TNBC, exploring its rationale and application across different disease stages. Trial registration NCT02555657.
    Keywords:  Clinical Trials; Immune checkpoint inhibitors; Immunotherapy; PDL-1; Triple-negative breast cancer; Tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1186/s43046-025-00295-x
  7. NPJ Precis Oncol. 2025 Jun 16. 9(1): 188
      Patients with high-grade serous ovarian cancer (HGSOC) typically present with widespread metastasis, obscuring a temporal understanding of tumor-immune dynamics. To address this, we perform multi-site global proteomics alongside matched immunohistochemistry (IHC) for CD4⁺ and CD8⁺ tumor-infiltrating lymphocytes (TILs) in patient samples. We order the protein expression profiles using an unbiased pseudotime analysis, recapitulating clinical observations of metastatic progression, and providing a framework to explore tumor-immune dynamics from localized to metastatic disease. Metastatic progression correlates with immune cell infiltration, the recruitment of regulatory T cells (Tregs) to counterbalance γδ T cell abundance, and an increased abundance of exhausted CD8⁺ T cells. The accumulation of Tregs at metastatic sites correlates with SNX8 expression, a critical regulator of the STING pathway. In early-stage tumors, keratin-expressing cancer cells recruit Tregs via MHC class II, fostering an inflammatory phenotype with limited IFNγ production and non-clonally expanded T cells. Together, our findings reveal novel mechanisms of immune escape associated with both localized disease and metastatic progression in HGSOC.
    DOI:  https://doi.org/10.1038/s41698-025-00973-y
  8. Ann Oncol. 2025 Jun 18. pii: S0923-7534(25)00806-3. [Epub ahead of print]
       BACKGROUD: To assess health-related quality of life (HRQoL) in patients with metastatic melanoma treated with a tumor infiltrating lymphocyte investigational medicinal product (TIL-IMP) or ipilimumab (IPI) after failure of first- or second-line treatment.
    PATIENTS AND METHODS: In a multicenter, randomized phase 3-trial, patients with metastatic melanoma (unresectable stage IIIC-IV) were randomized to TIL-IMP or IPI treatment (NCT02278887). HRQoL was measured using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire Core 15 Palliative Care (EORTC QLQ-C15-PAL), EuroQol 5D-3L (EQ-5D), and the Impact of Event Scale (IES). HRQoL outcomes were evaluated using a generalized estimating equations model. Sensitivity analyses were performed to assess drop-out assumptions. Results were interpreted for statistically and clinically significant differences.
    RESULTS: Of 168 patients randomized 1:1, n=143 baseline HRQoL scores (85%) were calculated. Of these, 75 (89%) were in the TIL-IMP-group and 73 (87%) in the IPI-group. TIL-IMP-treated patients reported higher global health status scores at week 24 compared to ipilimumab-treated patients (78.2 vs 73.9; p<0.05) and emotional functioning (85.9 vs 77.9; p<0.05). Patients treated with TIL-IMP had significantly lower fatigue (25.0 vs 32.4; p<0.05) and pain (13.7 vs 17.6; p<0.05) scores at week 24, but marginal higher nausea & vomiting (7.5 vs 5.2; p<0.05) symptom scores. EQ-5D scores were also higher in the TIL-IMP-group (0.89 vs. 0.83; p<0.05). These statistically significant differences were not considered clinically meaningful. The hypothesized higher impact of the more intensive TIL-IMP-treatment compared to IPI was not observed, according to IES scores (11.8 vs. 17.4; p<0.05 at 24 weeks). Sensitivity analyses showed similar results.
    CONCLUSIONS: Patients with metastatic melanoma treated with TIL-IMP experienced similar HRQoL compared to IPI monotherapy, with indications of better HRQoL outcomes over time. Ongoing follow-up is essential to assess longer-term HRQoL and its generalizability to a larger patient population to help guide personalized treatments decisions.
    Keywords:  Clinical Trial number: NCT02278887; Melanoma; cell therapy; immunotherapy; population-based; quality of life
    DOI:  https://doi.org/10.1016/j.annonc.2025.06.005
  9. Front Med (Lausanne). 2025 ;12 1555883
       Background: High-grade serous ovarian carcinoma (HGSOC), an aggressive cancer associated with pathogenic BRCA variants, causes genomic instability and sensitivity to poly (ADP-ribose) polymerase inhibitors. Identifying pathogenic BRCA variants is crucial for the treatment of HGSOC; however, genetic testing is expensive and time-consuming. This study aimed to explore pathological features, particularly the presence of tumor-infiltrating lymphocytes (TILs), as potential surrogates to streamline patient selection for genetic testing.
    Methods: We retrospectively analyzed 58 cases of HGSOC with known BRCA variant profiles. Tumors were categorized as TIL-positive or TIL-negative based on the presence of > 40 or ≤ 40 intraepithelial lymphocytes in a single high-power field (HPF), respectively. Key pathological features, including solid, endometrioid, and transitional (SET) architecture patterns; necrosis; and mitotic activity, were evaluated within these subgroups. Statistical analyses were used to determine the associations between these features and BRCA variant status.
    Results: In TIL-negative HGSOCs, SET patterns were strongly associated with pathogenic or likely pathogenic BRCA variants (p = 0.028), emerging as the most reliable morphological marker in this group. In TIL-positive HGSOCs, low mitotic activity (≤7 mitotic figure per 10 HPFs) was significantly correlated with pathogenic BRCA variants (p = 0.0002), underscoring its diagnostic significance. Necrosis and mitotic activity in TIL-negative cases and SET patterns in TIL-positive cases were not significantly associated with pathogenic BRCA variants. Combined analysis of both TIL subgroups diluted these associations, underscoring the significance of stratifying cases by the immune context.
    Discussion: The presence of TILs affects the diagnostic value of pathological features for BRCA variant status in HGSOC. Regarding pathogenic BRCA variants, SET patterns and low mitotic activity were identified as critical markers in TIL-negative tumors and TIL-positive tumors, respectively. These associations likely stem from interactions among genomic instability, immune response, and tumor growth. Our framework leverages these insights to prioritize high-risk cases for genetic testing, thereby optimizing resource allocation.
    Conclusion: The presence of TILs is critical for understanding the association between pathological features and pathogenic BRCA variants in HGSOC. To improve pathogenic BRCA variant prediction, optimize genetic testing, and guide tailored intervention, our framework integrates immune context and morphological markers. This approach is especially useful in resource-limited settings and can enhance diagnostic efficiency and clinical decision-making.
    Keywords:  BRCA; high-grade serous ovarian carcinoma; histology; mitosis; ovary; tumor infiltrating lymphocyte
    DOI:  https://doi.org/10.3389/fmed.2025.1555883
  10. Zhonghua Zhong Liu Za Zhi. 2025 Jun 23. 47(6): 508-516
      Objective: The impact of bystander CD8+ T cells (CD8+ Tbys) within the tumor microenvironment on the prognosis of early-stage non-small cell lung cancer (NSCLC) remains unclear, particularly concerning their infiltration differences at the invasive margin (IM) and tumor center (TC). Methods: We retrospectively collected postoperative specimens from 173 patients with primary early-stage NSCLC who underwent radical surgery at the Affiliated Tumor Hospital of Shandong First Medical University from 2014 to 2018. Tissue microarrays encompassing IM and TC regions were prepared and subjected to multicolor immunofluorescence staining (CK/CD8/CD103/DAPI). Image processing and phenotype recognition (CD8+ T cells, CK-CD8+; CD8+ Tbys, CK-CD8+CD103-) were performed using inFrom software, and automatic quantitative cell density analysis was conducted using R language. Differences in CD8+ T and CD8+ Tbys densities at IM and TC were analyzed using the Mann-Whitney U test. The relationship between CD8+ T, CD8+ Tbys and clinicopathological features was examined using the Kruskal-Wallis H test. The impact of CD8+ T and CD8+ Tbys on recurrence-free survival (RFS) was evaluated using Kaplan-Meier, log-rank, and Cox proportional hazards models. Results: A total of 173 patients with stage ⅠA-ⅡA NSCLC were included, with a recurrence rate of 26.6% (46/173) and a median RFS of 62.3 months (range: 44.7-71.9 months). CD8+ T and CD8+ Tbys densities (1/1 000) were significantly higher in the IM region than in the TC region [70(32, 155) vs. 37(18, 88), P<0.001; 25(11, 46) vs. 18(7, 34), P=0.002]. No significant association was found between CD8+ T, CD8+ Tbys and age, gender, smoking history, histological type, or pathological stage (all P>0.05). Patients with low-density IM CD8+ T cells had lower RFS compared to those with high-density IM CD8+ T cells (P=0.021; median RFS not reached), Further analysis revealed that patients with low-density IM CD8+ Tbys cell had lower RFS compared to those with high-density IM CD8+ Tbys (P=0.047; median RFS not reached), and low-density IM CD8+ T cell was an independent risk factor for postoperative recurrence (HR=1.836, 95% CI:1.007-3.347, P=0.048). Joint analysis of IM and TC revealed that patients with low IM CD8+ Tbys and high TC CD8+ Tbys had significantly lower RFS compared to the other three groups (P=0.006), and this combination was an independent risk factor for postoperative recurrence in early-stage NSCLC (HR=2.090, 95% CI:1.162-3.760, P=0.014). Conclusions: The spatial distribution of bystander CD8+ T cells within the primary tumor influences the prognosis of patients with early-stage NSCLC. Patients with low-density IM CD8+ Tbys and high-density TC CD8+ Tbys are more prone to recurrence after radical surgery.
    DOI:  https://doi.org/10.3760/cma.j.cn112152-20240805-00326
  11. Front Immunol. 2025 ;16 1576193
       Background: CD28+PD-1+ Tc cells (CD8+ T cells) constitute a dysfunctional subset of T cell; however, the mechanisms underlying their dysfunction and their significance in hepatocellular carcinoma (HCC) remain unclear. We aimed to elucidate the prognostic significance and molecular characteristics of CD28+PD-1+ Tc cell infiltration in HCC.
    Methods: We established a single-cell HCC transcriptional map, focusing on cell-cell communication and trajectory analysis of CD28+PD-1+ Tc cells. We assessed the correlation between CD28+PD-1+ Tc-cell enrichment and prognosis and investigated potential molecular mechanisms using enrichment analyses. Flow cytometry was used to compare CD28+PD-1+ Tc-cell infiltration between HCC and adjacent normal tissues and cytotoxic factors and immune checkpoint expression were evaluated.
    Results: Overall, 25,644 T cells were identified from single-cell RNA sequencing data from 10 HCC samples and corresponding normal samples. Overall T-cell infiltration was lower in HCC tissues, with significantly higher CD28+PD-1+ Tc-cell infiltration. Bulk RNA sequencing data integration revealed a correlation between higher CD28+PD-1+ Tc-cell infiltration and significantly worse prognosis. Flow cytometry confirmed higher CD28+PD-1+ Tc-cell enrichment in HCC tissues. Additionally, cytotoxic factor expression was significantly lower in CD28+PD-1+ Tc cells than in CD28-PD-1+ Tc cells, with lower expression of TIGIT and TIM-3 immune checkpoint molecules.
    Conclusions: Significantly high CD28+PD-1+ Tc-cell enrichment in HCC indicates potential immune dysfunction. CD28+PD-1+ Tc-cell enrichment may serve as a sensitive prognostic marker and indicator for predicting treatment responses.
    Keywords:  CD28; PD-1; hepatocellular carcinoma; single-cell RNA-seq; t cell exhaustion; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1576193
  12. Mol Oncol. 2025 Jun 15.
      Combination immunotherapies have entered the treatment armamentarium of oncology, but important knowledge gaps remain in our understanding of how these therapeutics work. A recent study by Rolig, Peng, and colleagues has shed new light on how dual blockade of PD1 and LAG3 enhances antitumor immunity. The authors first interrogated LAG3 expression on T cells across murine tumor models, classifying the models as LAG3hi or LAG3lo. Next, they found that LAG3hi models were unresponsive to anti-PD1 alone but responsive to combination therapy with anti-PD1 + anti-LAG3. Surprisingly, the response to anti-PD1 + anti-LAG3 in LAG3hi models was associated with reprogramming of CD4+ regulatory T cells (Treg) from the canonically immunosuppressive state to an inflammatory state characterized by loss of expression of the transcription factor Foxp3 and upregulation of transcription factor Tbet. Importantly, an analogous reprogrammed Treg state was associated with response to anti-PD1 + anti-LAG3 and longer overall survival in patients with metastatic melanoma. This work highlights the importance of cells beyond cytotoxic CD8+ T cells as drivers of response to immunotherapy and sets the stage for subsequent mechanistic and translational studies.
    Keywords:  Treg fragility; Treg stability; cancer immunology; immunotherapy; immuno‐oncology; regulatory T cells
    DOI:  https://doi.org/10.1002/1878-0261.70076
  13. Mol Ther Oncol. 2025 Jun 18. 33(2): 200996
      The increasing success of allogenic Vδ2 T cell immunotherapy for the treatment of cancer has been demonstrated in recent studies. Vδ2 T cells recognize phosphoantigens, intermediates of the mevalonate pathway, through butyrophilin molecules, and they are not major histocompatibility complex (MHC) restricted. Allogeneic transfer of in vitro expanded Vδ2 T cells has shown more promising results than autologous strategies, although the clinical benefit remains limited. One of the issues leading to less-than-optimal responses relates to the polyclonal expansion of Vδ2 T cell subsets with variable cytotoxic capacity. Previous work developed protocols to expand Vδ2 T cells, although to our knowledge, ours is the first comprehensive study that has produced a simple, antigen-presenting feeder-free culture that produced an average expansion of 3,000-fold and more than 95% pure Vδ2 T cells avoiding additional isolation steps. Here, we show the in vitro expansion of cytotoxic Vδ2 T cells expressing CD16 and NKG2A enriched in granzyme B that displayed enhanced antitumor activity of up to 40% against leukemia and ovarian, breast, and lung cancer cells. Our work warrants clinical testing to evaluate the therapeutic potential of these highly cytotoxic cells, paving the way for improved efficacy of personalized cell-based immunotherapies.
    Keywords:  CD137L; MT: Regular Issue; Vd2 cells; alendronate; cancer; cell therapy; cytotoxicity; expansion; gamma delta T cells; oncology
    DOI:  https://doi.org/10.1016/j.omton.2025.200996
  14. J Control Release. 2025 Jun 14. pii: S0168-3659(25)00580-2. [Epub ahead of print] 113960
      Mouse primary T cells have been engineered as a platform using chimeric antigen receptors (CARs) to induce the synthesis of desired proteins at the disease site. This approach allows for the use of immunocompetent syngeneic tumor models to evaluate the CAR T cells' function within the context of a fully functioning immune system. Current efforts to evaluate cell-based technologies typically rely on xenograft tumor models in immunodeficient mice, which provide early feasibility data but may not fully capture the immune effects present in the tumor microenvironment. In this study, a primary T-cell-based system for site-specific protein expression has been translated from human T cells to mouse T cells, allowing for the use of an immunocompetent syngeneic tumor model. A lentivector transduction, effective in human T cells, was adapted to engineer mouse T cells. CD4 and CD8 CAR T cell subsets were engineered separately and evaluated in immunocompetent mice for site-specific expression of the desired proteins. Co-expression of membrane-bound interleukin 15 (mbIL15) on the T cells enhanced intratumoral accumulation of both CD4 and CD8 CAR T cells and supported their delivery function. Validation of this platform in syngeneic models will enable efficacy assessments beyond solid tumors and allow for the evaluation of immune-related toxicities arising from interactions between the therapeutic protein, CAR T cells, and the host immune system."
    Keywords:  CAR T cells; Cell-based drug delivery; Immunocompetent mouse model; Interleukin-15; T-cell engineering; T-cell persistence
    DOI:  https://doi.org/10.1016/j.jconrel.2025.113960
  15. NPJ Vaccines. 2025 Jun 13. 10(1): 125
      TCR repertoires against tumors lack high-affinity TCRs and are further suppressed by Tregs. We hypothesized that Treg depletion enhances the antitumor efficacy of low-affinity T cells. Using the weak agonistic peptide A4Y derived from LCMV glycoprotein peptide p33 as a model antigen and VLPs as a vaccine platform, we tested this approach. In a separate low-affinity model, we targeted B16F10 melanoma with our multi-target vaccine. Results revealed limited in vivo lytic cross-reactivity between A4Y and p33 peptides, and the A4Y-vaccine alone failed to inhibit B16F10p33 tumor progression. However, combining A4Y-vaccine with Treg depletion triggered a robust immune response, characterized by increased CD8+ T cell infiltration, enhanced T cell functionality, and tumor-free survival. Infiltrating T cells also exhibited closer spatial proximity and heightened migration from blood vessels. Similarly, combining low-affinity vaccine with Treg depletion enhanced antitumor responses. These findings highlight the potential of Treg depletion to advance vaccination strategies targeting TAAs with low-affinity T cells.
    DOI:  https://doi.org/10.1038/s41541-025-01177-y
  16. Front Immunol. 2025 ;16 1609658
       Introduction: Although T-cell immunotherapies have been effective in the treatment of hematological malignancies, solid tumors have proven challenging due to the immunosuppressive microenvironment and lack of viable target antigens. The immune checkpoint ligand CD70, overexpressed in several solid tumors, yet with limited expression in healthy tissue, has emerged as a promising immunotherapeutic target.
    Method: This study describes the generation and preclinical characterization of ADP-520, a high-affinity, fratricide-resistant, CD70-targeted T-cell receptor fusion construct (TRuC) T-cell therapy enhanced with constitutively expressed mbIL-15, a membrane-bound fusion protein comprising interleukin-15 (IL-15) linked to full-length IL-15 receptor-alpha. The phenotypic distribution, expansion and persistence of ADP-520 TRuC T cells were measured in vitro under autonomous and antigen-dependent conditions, with the contributions of TCR and IL-15 signaling pathways ascertained using inhibition assays. Chronic antigen stimulation was used to evaluate exhaustion-resistance, while anti-tumor potency was explored both in vitro and in vivo.
    Results: ADP-520 was found to have potent and antigen-specific activity against hematological and solid CD70-expressing tumors, without apparent fratricide or killing of bystander T cells despite CD70 expression by activated lymphocytes. Engineered co-expression of mbIL-15 augmented antigen-dependent expansion through pro-survival effects and enrichment of an early memory T-cell phenotype, thus enhancing tumor-autonomous, exogenous cytokine-free persistence and bolstering exhaustion resistance during chronic stimulation. mbIL-15 co-expression also enhanced intratumoral T-cell infiltration in vivo for potent and persistent antitumor efficacy.
    Discussion: These findings characterize ADP-520 as a first-in-class, CD70-targeted, fratricide-resistant autologous TRuC T-cell therapy leveraging native TCR signaling combined with constitutive IL-15 signaling to impart T cells with enhanced persistence, tumor penetration, and antitumor efficacy. This makes ADP-520 a promising cell immunotherapy candidate for clinical development, with the potential to overcome hurdles intrinsic to the treatment of solid tumors.
    Keywords:  CD70; IL-15; T-cell immunotherapy; TRuC T-cell therapy; fratricide resistant; persistence; renal cell carcinoma; solid tumor
    DOI:  https://doi.org/10.3389/fimmu.2025.1609658
  17. Int Immunopharmacol. 2025 Jun 13. pii: S1567-5769(25)01022-7. [Epub ahead of print]161 115032
      B7-H3, an immune-checkpoint molecule that is overexpressed in several cancer types, has been identified as a promising immunotherapy target. However, most immunotherapy approaches against B7-H3+ tumor cells focus on manipulating the T cells. Natural killer (NK) cells, another important part of the cellular immune system, also exhibit anti-tumor properties and play complementary roles in tumor eradication with T cells. In this study, natural killer group 2D (NKG2D), an activating receptor in most cytotoxic immune cells, was selected for engaging NK cells. We obtained specific anti-NKG2D nanobodies via phage display and developed a series of B7-H3 × NKG2D bispecific antibodies (bsAb) with different formats to fight against the B7-H3+ tumor cells. Through functional comparison of candidate antibodies in vitro, B1-C53 was selected and further modified with the optimized Fc fragment (known as FC-C53) to enhance anti-tumor immunity by antibody-dependent cell-mediated cytotoxicity (ADCC). Stronger tumor lysis mediated by FC-C53 was demonstrated both in vitro and in vivo when simultaneously directed at both NK cells and CD8+ T cells, particularly after the additional administration of a B7-H3 × CD3 bispecific T cell engager that targets B7-H3 with another epitope. Overall, we provided a strategy based on the B7-H3 × NKG2D antibody to improve the anti-B7-H3 immunotherapy approaches by orchestrating cytotoxic lymphocytes.
    Keywords:  ADCC; B7-H3; Bispecific antibody; NK cells; NKG2D; Nanobody; Synergistic effect
    DOI:  https://doi.org/10.1016/j.intimp.2025.115032