bims-tuinly Biomed News
on Tumor-infiltrating lymphocytes therapy
Issue of 2025–08–03
fifteen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Cancers (Basel). 2025 Jul 08. pii: 2269. [Epub ahead of print]17(14):
      Background/Objectives: Tumor-infiltrating lymphocytes (TILs) and inflammation status are emerging prognostic markers in various cancers, but their significance in high-grade serous ovarian carcinoma (HGSC) remains unclear. Our objective was to evaluate different TIL subtypes and inflammation status in relation to progression-free survival (PFS) in primary HGSC. Methods: CD3+/CD4+/CD8+/PD-1+ stromal TILs (sTILs) and intraepithelial TILs (iTILs) were evaluated by manual assessment and digital image analysis (DIA), following TIL Working Group recommendations. Inflammation status was evaluated through the following scores: systemic immune-inflammation index (SII), pan-immune-inflammation value (PIV), CA125, and lactate dehydrogenase (LDH). Results: CD8+ TILs were the most prevalent subtype in both iTILs and sTILs. However, sTILs were significantly more abundant than iTILs (p < 0.001) among all subsets, except for PD-1+ cells. DIA results of TIL assessments were in agreement with manual assessments. High stromal CD3+ and CD8+ TILs, PIV, CA125, and LDH, were associated with improved PFS. Potential independent prognostic factors for PFS in manual assessment were PIV (HR = 0.32, CI 95% = 0.12-0.82) and CD8+ sTILs (HR = 0.30, CI 95% = 0.12-0.79), whereas in DIA assessment they were CD3+ sTILs (HR = 0.31, CI 95% = 0.15-0.67), PIV (HR = 0.35, 95% CI 0.13-0.96), and residual disease (HR = 0.21 95% CI 0.08-0.53). Conclusions: CD3+/CD8+ sTILs and PIV are promising prognostic indicators in HGSC; however, further research is needed to confirm their clinical utility.
    Keywords:  digital-image analysis; high-grade serous ovarian carcinoma; immunohistochemistry; inflammation status; manual scoring; prognostic biomarkers; tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.3390/cancers17142269
  2. J Transl Med. 2025 Jul 25. 23(1): 842
       BACKGROUND: Lymph node metastasis (LNM) is critical for staging, prognosis, and treatment decisions in lung adenocarcinoma (LUAD). While tumor-infiltrating lymphocytes (TILs) have demonstrated prognostic value, their role in LNM risk remains uninvestigated. This study evaluates the relationship between TIL features from primary tumor whole slide images (WSIs) and LNM in LUAD.
    METHODS: TILScout was utilized to derive patch-level TIL scores and generate global TIL maps from primary tumor WSIs. Hot spot analysis and deep learning-based feature extraction followed by K-means clustering were applied to identify and characterize spatial TIL clusters (sTILCs) from the global TIL maps. Random forest models incorporating clinical/pathological data with (M1) and without (M2) TIL features (TIL scores and sTILCs) were developed on a training cohort (N = 312) to predict LNM, and performance was compared across validation (N = 78) and independent test cohorts (N = 148).
    RESULTS: Two sTILC types ("TIL-cold" cluster [sTILC1] and "TIL-hot" cluster [sTILC2]) were identified. Model M1 significantly improved LNM prediction over M2, with AUCs increasing from 0.63 to 0.78 (Z = 5.366, P < 0.001) and from 0.61 to 0.72 (Z = 1.999, P = 0.046) in the training and validation cohorts, and from 0.69 to 0.80 (Z = 3.030, P = 0.002) in the test cohort. Decision curve analysis indicated that M1 provided greater net benefit across a broad spectrum of threshold probabilities. Importantly, patients with lower TIL scores and/or classified as sTILC1 consistently had an increased risk of LNM.
    CONCLUSIONS: Spatial TIL features in primary tumors are linked to LNM in LUAD, thereby enabling the identification of high-risk patients and guiding personalized treatment strategies.
    Keywords:  Lung adenocarcinoma; Lymph node metastasis; TILScout; Tumor-infiltrating lymphocytes; Whole slide images
    DOI:  https://doi.org/10.1186/s12967-025-06860-1
  3. Int J Mol Sci. 2025 Jul 15. pii: 6778. [Epub ahead of print]26(14):
      Cutaneous melanoma is an aggressive cancer with an increasing incidence worldwide, highlighting the need for research into its pathogenesis. The tumor microenvironment (TME) plays a critical role in melanoma progression and consists of cellular components and an extracellular matrix (ECM) rich in cytokines and signaling molecules. The most abundant stromal cells within the TME are cancer-associated fibroblasts (CAFs), which remodel the ECM and modulate immune responses. Among immune cells, tumor-associated macrophages (TAMs) predominate, and their polarization toward the M2 phenotype supports tumor progression. Tumor-infiltrating lymphocytes (TILs) have diverse functions, including cytotoxic T-cells, helper T-cells that modulate immune response, B-cells forming tertiary lymphoid structures (TLS), and regulatory T-cells with immunosuppressive properties. Dendritic cells (DCs) also play a complex role in the TME. A notable subpopulation are mature regulatory dendritic cells (mregDCs), which contribute to immune evasion. All of these TME components may drive tumorigenesis. Advancements in melanoma treatment-including immunotherapy and targeted therapies-have significantly improved outcomes in advanced-stage disease. In parallel, emerging approaches targeting the tumor microenvironment and gut microbiome, as well as personalized strategies such as neoantigen vaccines and cell-based therapies, are under active investigation and may further enhance therapeutic efficacy in the near future.
    Keywords:  cancer/melanoma-associated fibroblasts (CAFs); dendritic cells; immune checkpoint inhibitors (ICI); macrophages; melanoma; metastases; tumor microenvironment; tumor-infiltrating lymphocytes (TILs)
    DOI:  https://doi.org/10.3390/ijms26146778
  4. J Hepatocell Carcinoma. 2025 ;12 1633-1645
       Background: HHLA2, a member of the B7 family, is extensively expressed in various cancers and plays a pivotal role in modulating the immune microenvironment. However, its prognostic significance in hepatocellular carcinoma (HCC) remains poorly understood. This study aims to elucidate the expression patterns of HHLA2 and PD-L1 in HCC, their associations with tumor-infiltrating lymphocytes (TILs), and their impact on clinical outcomes.
    Methods: Immunohistochemistry (IHC) was employed to evaluate HHLA2 and PD-L1 expression in 547 HCC tissue samples. PD-L1 positivity was defined as ≥1% membranous or cytoplasmic staining. Hematoxylin and eosin (H&E) staining was utilized to quantify TILs (percentage/area), while IHC was used to measure the densities of CD3+, CD4+, and CD8+ TILs (cells/mm²).
    Results: HHLA2 and PD-L1 exhibited similar positivity rates. HHLA2 positivity was associated with older age, lower alpha-fetoprotein (AFP) levels, well-differentiated tumors, and improved overall survival (OS). HHLA2 expression was inversely correlated with stromal TIL density. In contrast, tumor cell (TC)-PD-L1 and inflammatory cell (IC)-PD-L1 positivity were positively correlated with higher stromal TIL density and increased levels of CD3+, CD4+, and CD8+ TILs. Patients with HHLA2(+)/PD-L1(-) status demonstrated the longest OS. A novel classification system based on HHLA2/PD-L1 expression identified distinct immune profiles and prognostic subgroups.
    Conclusion: HHLA2 significantly influences the immune microenvironment of HCC and serves as an independent prognostic marker. The combined assessment of HHLA2 and PD-L1 expression facilitates risk stratification, providing a framework to optimize immunotherapy strategies. These findings contribute to the advancement of precision medicine in the management of HCC.
    Keywords:  HCC; HHLA2; PD-L1; TILs; prognosis
    DOI:  https://doi.org/10.2147/JHC.S513033
  5. Acad Radiol. 2025 Jul 29. pii: S1076-6332(25)00656-7. [Epub ahead of print]
       RATIONALE AND OBJECTIVES: To develop and validate a nomogram based on clinicopathologic features, conventional ultrasound and dual-mode elastography for predicting breast cancers (BCs) with high level of tumor-infiltrating lymphocytes (TILs).
    METHODS: A total of 520 patients with 527 invasive breast cancers (cT1or2, N negative or positive, M0) were enrolled retrospectively from January 2023 through November 2023. All patients underwent conventional ultrasound and dual-mode elastography (strain elastography, SE, and shear wave elastography. SWE). Stiffness values of both the entire tumor and the surrounding tissues were recorded. A nomogram was developed by using logistic regression and determined by receiver operating characteristic (ROC) curve analysis and calibration curve analysis.
    RESULTS: Among the 527 lesions enrolled into this study,83.3% (439/527) were tumors with low-moderate TILs and 88 (16.7%) with high TILs. ER/PR negative, high histologic grade, LPBC-associated shape and posterior acoustic enhancement on conventional ultrasound, decreased FLR on SE, lower Shell2mm Emean on SWV were significantly associated with high TILs BCs. A nomogram constructed with the above six variables enabled predicting tumors with high TILs in the training cohort (AUC=0.874) and internal validation cohort (AUC=0.913). The nomogram also showed good calibration in the training cohort and internal validation cohorts (Hosmer Lemeshow goodness-of-fit test, p = 0.824 and p = 0.536, respectively).
    CONCLUSION: High TILs breast cancers had specific clinicopathological features, benignlike morphological characteristics on conventional ultrasound and lower stiffness on elastography. A preoperative nomogram integrating the above features was useful in predicting high TILs tumors, which might help individualized management of tumors.
    Keywords:  Elastography; Tumor-infiltrating lymphocyte; Ultrasound; breast cancer
    DOI:  https://doi.org/10.1016/j.acra.2025.07.016
  6. Clin Transl Radiat Oncol. 2025 Sep;54 101018
       Background: Antitumor immunity, exerted by CD8+ cytotoxic T lymphocytes, plays a vital role in tumor control. Therefore, the present study was conducted to compare the amount of CD8+ tumor-infiltrating lymphocytes (TILs) before and after either long- (LCRT) or short-course radiotherapy (SCRT) in rectal cancer.
    Methods: This study retrospectively assessed rectal cancer patients treated by neoadjuvant radiotherapy between 2019 and 2021. Biopsy and surgical samples were subjected to immunohistochemical staining to count CD8+ TILs. The association between the post-to-pre-treatment CD8+ count ratio and treatment groups, histopathological factors, and response to treatment was assessed.
    Results: A total of 34 patients were included, with 23 (67.6 %) receiving LCRT and 11 (32.4 %) receiving SCRT. The mean age was 58.56 ± 13.59 years. The number and percentage of CD8+ TILs increased significantly after radiotherapy in all patients (P < 0.001). An increase in CD8+ TILs was observed in both groups, with LCRT showing a median post-to-pre-treatment count ratio of 2.77 and SCRT showing 3.1 (P = 0.127). A generalized linear multivariate model adjusting for mucinous histology, surgical grade, and pathological stages revealed that SCRT was associated with a significantly higher post-to-pre-treatment CD8+ count ratio compared to LCRT (P = 0.03).
    Conclusion: Our study highlights that both SCRT and LCRT significantly increase CD8+ TIL count and percentage, reflecting robust immune activation after radiotherapy in rectal cancer, with SCRT showing a higher relative increase, though not statistically significant in unadjusted analyses. After adjusting for histopathological variables, SCRT was independently associated with a greater increase in CD8+ T cells.
    Keywords:  Neoadjuvant radiotherapy; Rectal cancer; Tumor-infiltrating CD8-positive lymphocytes
    DOI:  https://doi.org/10.1016/j.ctro.2025.101018
  7. Cancer Cell. 2025 Jul 22. pii: S1535-6108(25)00276-4. [Epub ahead of print]
      Immunotherapy has shown limited success in recurrent ovarian cancer (OC), with prognostic insights largely derived from treatment-naive tumors. We analyzed 697 tumor samples (566 primary and 131 recurrent) from 595 OC patients across five independent cohorts, capturing tumor-infiltrating lymphocytes (TILs) heterogeneity and identifying four immune phenotypes linked to prognosis and TIL:myeloid networks driving malignant progression. We found that in preclinical mouse models, mirroring inflamed human OCs, the recurrent Brca1mut tumors maintained activated TILs:dendritic cells (DCs) niches but evaded immune control through upregulation of COX/PGE2 signaling. Conversely, recurrent Brca1wt tumors displayed loss of TILs:DCs niches and accumulated immunosuppressive tumor microenvironment (TME) networks featuring Trem2/ApoEhigh tumor associated macrophages (TAMs) and Nduf4l2high/Galectin3high malignant states. Recurrent tumors recapitulate the immunogenic landscapes of original cancers. Our findings reveal BRCA-dependent TIL:myeloid crosstalk as key to persistent immunogenicity in recurrent OC and propose new targets to enhance chemotherapy efficacy.
    Keywords:  PGE(2) signaling; TREM2; immune phenotype; myeloid-T cell networks; ovarian cancer; recurrence; tumor microenvironment
    DOI:  https://doi.org/10.1016/j.ccell.2025.07.005
  8. Cell Rep Med. 2025 Jul 24. pii: S2666-3791(25)00325-8. [Epub ahead of print] 102252
      Most breast cancers express the estrogen receptor (ER), but the immune response of hormone receptor-positive (HR+) breast cancer remains poorly characterized. Here, dendritic cells loaded with tumor lysate are used to identify tumor-reactive CD8 T cells, which are detected in most HR+ breast cancer patients, especially those with early-stage tumors. When present, the circulating antitumor CD8 response contains cytotoxic T cells with diverse specificity and T cell receptor (TCR) repertoire. Additionally, patients with blood cancer-specific T cells have significantly more CD8 tumor-infiltrating lymphocytes (TILs). Moreover, tumor-reactive TCR sequences are detected in the tumor, but at a significantly lower proportion in patients with lymph node involvement. Our data suggest that HR+ breast cancer patients with lymph node metastasis lack tumor-specific CD8 T cells with capacity to infiltrate the tumor at significant levels. However, early-stage patients have a diverse antitumor CD8 response that could be harnessed to develop immunotherapeutic approaches for late-stage HR+ patients.
    Keywords:  CDR3; CTA; ER+ breast cancer; T cell receptor; T cell repertoire; antitumor CD8 T cells; cancer testis antigen; metastatic luminal breast cancer; moDC; tumor lysate; tumor-specific T cells
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102252
  9. Discov Oncol. 2025 Jul 28. 16(1): 1435
      With the widespread use of mammography, the incidence of ductal carcinoma in situ (DCIS) has significantly increased and currently accounts for approximately 20-25% of newly diagnosed breast cancer cases. While a significant proportion of DCIS lesions exhibit long-term indolence without progression to invasive carcinoma, the current paucity of validated biomarkers for invasive potential prediction creates a therapeutic dilemma in clinical practice, potentially leading to either overtreatment of biologically inert lesions or undertreatment of high-risk precursors. Reflecting the body's adaptive immune response, tumor-infiltrating lymphocytes (TILs) have been demonstrated to be associated with adverse clinicopathologic factors in DCIS. The relationship between total TILs and the risk of DCIS recurrence remains controversial, but subpopulations of TILs such as FOXP3 + T lymphocytes, B cells, CD68, and CD163 macrophages are found to be associated with DCIS recurrence. Increased levels of total TILs are associated with increased malignancy of breast lesions, and the number and composition of TILs subpopulations change during DCIS progression. Moreover, dense TILs are correlated with high PD-L1 expression and the "healing" phenomenon in DCIS. Further exploration of the immune microenvironment, TILs subpopulations, and adaptive immune responses in DCIS remains a critical unmet need, contributing to more refined risk stratification of DCIS patients and facilitating the implementation of new treatments.
    Keywords:  Adaptive immunity; Ductal carcinoma in situ; Immune checkpoint; Immune microenvironment; Immunotherapy; Prognosis; Tumor-infiltrating lymphocytes
    DOI:  https://doi.org/10.1007/s12672-025-03288-3
  10. Cancer Res Treat. 2025 Jul 28.
       Purpose: Immune excluded/desert tumors show reduced responsiveness to immunotherapy compared to inflamed tumors. The tumor stroma contributes to immune evasion. This study aimed to identify proteins overexpressed in tumors with high tumor stroma among immune excluded triple-negative breast cancer (TNBC) to better understand the mechanisms of immune exclusion.
    Materials and Methods: Proteomic analysis was conducted on formalin-fixed, paraffin-embedded samples from 403 cases of TNBC. We compared protein expression between stroma high vs. stroma low within the immune excluded subtype. We investigated the correlations between the identified protein expression and other clinicopathologic features. Immunohistochemical (IHC) staining and single-cell analysis were conducted, along with survival analysis.
    Results: Among the 247 eligible cases, 81 (32.8%) were classified as immune excluded and 166 (67.2%) as inflamed. Within the excluded subtype, periostin was the only extracellular matrix-related protein significantly overexpressed in stroma high cases. Periostin expression demonstrated a positive correlation with the amount of stroma (r = 0.51, p<0.001) and a negative correlation with tumor-infiltrating lymphocytes (TILs) (r = -0.30, p<0.001). Periostin expression in the tumor stroma was confirmed by IHC. Single-cell analysis demonstrated that periostin originated from cancer-associated fibroblasts (CAFs). High periostin levels correlated with poorer recurrence-free survival (hazard ratio 1.422, p=0.005).
    Conclusion: Periostin is overexpressed in stroma high, immune excluded TNBC and is derived from CAFs. Its expression is associated with reduced TILs and poor prognosis. The development of targeted agents against periostin-positive CAFs may help overcome immune evasion and improve the effectiveness of immunotherapy in TNBC.
    Keywords:  Immune subtypes; Periostin; Proteomic analysis; Triple-negative breast neoplasms; Tumor stroma
    DOI:  https://doi.org/10.4143/crt.2025.145
  11. Front Immunol. 2025 ;16 1626085
      Regulatory T cells (Tregs) are a small, unique subset of suppressive T cells that play a pivotal role in regulating the immune system by maintaining tolerance to self-antigens and preventing autoimmune disease. Adoptive transfer of Tregs for the treatment of autoimmune disorders such as arthritis and allergic airway inflammation, graft-versus-host disease (GvHD) and rejection following transplant have shown promise in early phase clinical trials. Despite over a decade of clinical manufacturing, there remains significant manufacturing and testing complexities for this class of therapies, including the need for specialized facilities and highly trained personnel that make clinical and commercial supply challenging. In this review, we discuss the current Chemistry, Manufacturing and Controls (CMC) and regulatory complexities and challenges to the development and commercialization of Treg therapies. Some of these are specific to Tregs while others are broadly applicable to the field of cell-based therapy. Discussion topics include the importance of starting material selection, the availability of GMP quality reagents and material, isolation and characterization of regulatory T cells, cGMP manufacturing considerations and limitations, the complexity of testing, release and distribution of cell-based therapies, as well as the regulatory challenges associated with Treg therapy. Treg cell therapies can be fraught with technical challenges which are mirrored by a sponsor's ability to meet regulatory requirements. Despite these hurdles, the promise of Tregs as a therapeutic for the treatment of autoimmune and other diseases warrants continued development.
    Keywords:  ACT; CMC; Treg; analytics; manufacturing; regulatory
    DOI:  https://doi.org/10.3389/fimmu.2025.1626085
  12. Clin Cancer Res. 2025 Aug 01.
       PURPOSE: T cell stemness is important for anti-tumor immunity. The presence in tumor draining lymph node and tumor of stem-like memory T cells (TSCM) and T cells expressing the transcription factor TCF1, critical in T cell self-renewal, are associated with improved response to immune checkpoint inhibitors.
    METHODS: We studied the effects of the tumor-targeting immunocytokine PDS01ADC (NHS-IL12) on peripheral T cell stemness in murine hosts and in patients with advanced solid tumors. TSCM and expression of the murine T cell stemness markers SCA1 (Ly6a) and Tcf7 were evaluated in naïve and tumor-bearing mice receiving murine PDS01ADC (NHS-muIL12). Peripheral blood from patients treated in a clinical trial with PDS01ADC was analyzed for TSCM and expression of TCF1 on T cell subsets.
    RESULTS: NHS-muIL12 treatment in naïve mice increased SCA1+ peripheral T cells with stem-like phenotypes and promoted tumor infiltration of CD8+ T cells displaying increased stemness. In patients with advanced solid tumors, PDS01ADC treatment increased peripheral CD8+ and CD4+ TSCM frequencies and effector memory T cells expressing TCF1, with greater increases associated with disease control. Most peripheral TSCM were negative for PD-1 and TIGIT throughout treatment, suggesting their quiescence and self-renewal capacity. Expanded TCF1-negative effector memory CD8+ T cells expressed PD-1 with increased intensity of granzyme B, indicating an activated, cytotoxic state.
    CONCLUSIONS: PDS01ADC treatment in patients with advanced solid tumors boosts peripheral T cells with stem-like characteristics, correlating with disease stabilization. Further studies combining PDS01ADC with other immunotherapies to synergize with this peripheral burst of T cell stemness are warranted.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-25-1490
  13. Front Immunol. 2025 ;16 1618393
      Unlike conventional T cells that detect peptide antigens loaded to major histocompatibility complex (MHC) molecules, unconventional T cells respond to non-peptidic metabolite antigens presented by MHC class I-like proteins, such as CD1 and MHC-related protein 1 (MR1). Semi-invariant mucosal-associated invariant T (MAIT) cells, γδ T cells, and invariant natural killer T (iNKT) cells, together with other CD1- or MR1-restricted T cell subsets expressing diverse T cell receptors (TCR), elicit an innate-like response independent of diverse MHC genetics. In contrast to an overall enhanced response to bacterial-derived riboflavin precursor metabolites in infections, MAIT cells often exhibit an immunosuppressive or exhausted phenotype in glioblastoma, lung cancer, colorectal cancer, and various hematological malignancies. Whereas some tumor cells can activate MAIT cells, the structures and functions of tumor-derived MR1 ligands remain largely unknown. Novel discoveries of mammalian-derived agonists and antagonists binding to MR1 protein are our knowledge of MR1 ligand structures and functions from MAIT cell activation in healthy conditions to anti-cancer immunity. Recent findings reveal that nucleoside and nucleobase analogs, as self-metabolites to activate MR1-restricted T cells, are regulated in the tumor microenvironment. Likewise, iNKT cells exhibit a dynamic role in cancer, capable of both protumor and antitumor immunity. Similarly, γδ T cells have also demonstrated both protective and tumor-promoting roles, via recognizing stress-induced protein and metabolite ligands. This review further depicts the distinct kinetics of responses, highlighting a rapid activation of unconventional T cells in solid versus hematological cancers. Emerging therapeutic strategies, including antigen-loaded MR1 and CD1, adoptive T cell transfer, chimeric antigen receptor-T (CAR-T) cells, T cell receptor-T (TCR-T) cells, and combination treatments with immune checkpoint inhibitors, yet remain challenging, hold promise in overcoming tumor-induced immunosuppression and genetic restriction of conventional T cell therapies. By addressing critical gaps, such as novel structures and functions of cancer metabolite antigens, unconventional T cells offer unique advantages in anti-cancer immunotherapy.
    Keywords:  CD1; MHC class I-related protein 1 (MR1); cancer; immunotherapy; lipids; polar metabolites; unconventional T cells
    DOI:  https://doi.org/10.3389/fimmu.2025.1618393
  14. Nat Immunol. 2025 Aug;26(8): 1367-1383
      CD8+ T cell-mediated tumor control and efficacy of immune checkpoint blockade (ICB) are associated with both precursors of exhausted T (TPEX) cells and tissue-resident memory T cells. Their relationships and relative contribution to tumor control, however, are insufficiently understood. Using single-cell RNA sequencing and genetic mouse models, we systematically dissected the heterogeneity and function of cytotoxic T cells in tumors and tumor-draining lymph nodes (tdLNs). We found that intratumoral TCF1+ TPEX cells and their progeny acquired a tissue-residency program that limits their contribution to tumor control and ICB response. By contrast, MYB-dependent stem-like TPEX cells residing in tdLNs sustained CD8+ T cell infiltration into tumors and mediated ICB response. The cytokine TGFβ was the central factor that enforced residency of intratumoral CD8+ T cells and limited the abundance of stem-like TPEX cells in tdLNs, thereby restraining tumor control. A similar network of TGFβ-constrained intratumoral and extratumoral CD8+ T cells with precursor and residency characteristics was found in human cancer.
    DOI:  https://doi.org/10.1038/s41590-025-02219-2
  15. J Immunother Cancer. 2025 Jul 31. pii: e012029. [Epub ahead of print]13(7):
       BACKGROUND: T-cell receptor (TCR)-T immunotherapy has emerged as a promising strategy for cancer treatment. However, identifying TCRs that can be used to generate TCR-T cells remains challenging due to tumor heterogeneity, the scarcity of tumor-specific T cells, and the diversity of human leukocyte antigens (HLA). To advance TCR-T immunotherapy, it is crucial to develop an efficient and scalable method to identify tumor-specific TCRs.
    METHODS: To identify tumor-specific TCRs, epitopes, and their corresponding HLA subtypes, we developed a method for rapidly assembling TCRs identified through the single-cell analysis of T cells from various tumors. For each TCR, only two pairs of oligonucleotides corresponding to the CDR3 regions of TCR-α and β chains needed to be synthesized, enabling the construction of a TCR library quickly in a cost-effective manner. Additionally, we engineered HLA-knockout HEK-293T cells as antigen-presenting cells to express patient-specific HLA-I and neoantigens, and a Jurkat NFAT-GFP reporter cell line for screening antigen-reactive TCRs. The efficacy of our TCR-screening system was validated through a small-scale screening of HPV16-specific TCRs from patients with cervical cancer.
    RESULTS: We successfully developed a TCR assembly method that enables the rapid cloning and construction of TCR libraries within 2 days, significantly accelerating the process and reducing costs. Our antigen-presenting system also allows for flexible expression of patient-specific HLA-I molecules, facilitating personalized screening. The Jurkat reporter cells demonstrated high sensitivity for screening functional TCRs. Using published datasets from patients with HPV16-positive cervical cancer, we successfully used our system to isolate a human papillomavirus (HPV)-specific TCR. Through deletion, alanine scanning, and mass spectrometry analysis, we determined that this TCR specifically recognized an 8-mer peptide (MHGDTPTL) from HPV-E7 presented by HLA-B*15:18. Moreover, T cells expressing this TCR were able to effectively kill HPV-positive cells.
    CONCLUSIONS: We developed an integrated antigen-presenting, TCR assembly, and TCR reporter system for screening tumor-specific TCRs using single-cell sequencing datasets. By using this system, we have successfully identified a functional, HPV-specific TCR, demonstrating the potential of our approach for the efficient screening of tumor-specific TCRs to advance TCR-T immunotherapy.
    Keywords:  Adoptive cell therapy - ACT; Cervical Cancer; Human leukocyte antigen - HLA; T cell Receptor - TCR
    DOI:  https://doi.org/10.1136/jitc-2025-012029