Pub Date : 2025-12-04DOI: 10.1136/jitc-2025-013024
Monika Sponheimer, Kieron White, Michelle Ulrich, Maryam Kazerani, Andreas Maiser, Emil Thore Tyborski, Giulia Petra Rappa, Daniel Richter, Eva Briem, Gerulf Hänel, Nora Philipp, Daniel Nixdorf, Lisa Rohrbacher, Anetta Marcinek, Andreas Linder, Niklas Kuhl, Ignazio Piseddu, Tobias Straub, Hartmann Harz, Christian Wichmann, Denis Maenner, Martina Rudelius, Veit Hornung, Heinrich Leonhardt, Catharina H M J Van Elssen, Sherif Abdelhamed, Daniel Diolaiti, Veit Bücklein, Marion Subklewe
Background: Challenges to developing immunotherapies for acute myeloid leukemia (AML) include the identification of suitable target antigens due to on-target-off-leukemia toxicity. CD70, expressed on AML bulk and leukemic stem cells with limited expression on healthy cells, has emerged as a promising target.
Methods: This study evaluated CD70 as a target for NK-cell-based immunotherapy using a sugar-engineered antibody (PF-08046040, SEA-CD70). CD70 surface expression was assessed in primary AML samples by multiparameter flow cytometry. The cytotoxic capacity of SEA-CD70 was analyzed through antibody-dependent cellular cytotoxicity (ADCC) assays using AML cell lines, primary AML samples, and a severe combined immunodeficiency (SCID) mouse xenograft model. The effects of cytokines on CD70 expression and ADCC were investigated by exposing AML cells to conditioned medium (CM) derived from activated T cells or recombinant cytokines.
Results: Flow cytometry revealed CD70 expression ranging from 0.2% to 89.6% (median=7.0%, n=86) in primary AML cells across genetic subgroups; this expression remained unchanged at relapse (median=3.9%, n=14). SEA-CD70 showed potent, dose-dependent cytotoxicity against AML cell lines, primary cells, and in an SCID mouse model, which correlated with CD70 expression levels. Notably, AML cells exposed to CM from activated T cells upregulated CD70. TNF-α was identified as the driver of CD70 upregulation, translating into enhanced ADCC against AML cells (cytotoxicity w/o TNF-α = 17.9% vs with TNF-α = 34.3%, n=13-15). Conversely, IFN-γ exposure led to reduced ADCC (cytotoxicity w/o IFN-γ = 17.9% vs with IFN-γ = 9.2%, n=15), which is attributed to increased expression of NK inhibitory receptor ligands (HLA-ABC, HLA-E). Blocking of the corresponding inhibitory NK receptors (KIR/CD158b and NKG2A) partially reversed this effect. Similar findings were observed with a CD33-directed antibody, indicating a universal resistance mechanism against ADCC-based immunotherapy in AML.
Conclusions: CD70 is a promising target for NK cell-based immunotherapy in AML. However, IFN-γ-dependent upregulation of HLA molecules on AML cells contributes to resistance to ADCC. These findings underscore the need for rationale combination strategies in clinical trials to overcome this inducible immune escape mechanism.
{"title":"TNF-α and IFN-γ differentially regulate AML cell susceptibility to CD70-antibody-mediated cytotoxicity.","authors":"Monika Sponheimer, Kieron White, Michelle Ulrich, Maryam Kazerani, Andreas Maiser, Emil Thore Tyborski, Giulia Petra Rappa, Daniel Richter, Eva Briem, Gerulf Hänel, Nora Philipp, Daniel Nixdorf, Lisa Rohrbacher, Anetta Marcinek, Andreas Linder, Niklas Kuhl, Ignazio Piseddu, Tobias Straub, Hartmann Harz, Christian Wichmann, Denis Maenner, Martina Rudelius, Veit Hornung, Heinrich Leonhardt, Catharina H M J Van Elssen, Sherif Abdelhamed, Daniel Diolaiti, Veit Bücklein, Marion Subklewe","doi":"10.1136/jitc-2025-013024","DOIUrl":"10.1136/jitc-2025-013024","url":null,"abstract":"<p><strong>Background: </strong>Challenges to developing immunotherapies for acute myeloid leukemia (AML) include the identification of suitable target antigens due to on-target-off-leukemia toxicity. CD70, expressed on AML bulk and leukemic stem cells with limited expression on healthy cells, has emerged as a promising target.</p><p><strong>Methods: </strong>This study evaluated CD70 as a target for NK-cell-based immunotherapy using a sugar-engineered antibody (PF-08046040, SEA-CD70). CD70 surface expression was assessed in primary AML samples by multiparameter flow cytometry. The cytotoxic capacity of SEA-CD70 was analyzed through antibody-dependent cellular cytotoxicity (ADCC) assays using AML cell lines, primary AML samples, and a severe combined immunodeficiency (SCID) mouse xenograft model. The effects of cytokines on CD70 expression and ADCC were investigated by exposing AML cells to conditioned medium (CM) derived from activated T cells or recombinant cytokines.</p><p><strong>Results: </strong>Flow cytometry revealed CD70 expression ranging from 0.2% to 89.6% (median=7.0%, n=86) in primary AML cells across genetic subgroups; this expression remained unchanged at relapse (median=3.9%, n=14). SEA-CD70 showed potent, dose-dependent cytotoxicity against AML cell lines, primary cells, and in an SCID mouse model, which correlated with CD70 expression levels. Notably, AML cells exposed to CM from activated T cells upregulated CD70. TNF-α was identified as the driver of CD70 upregulation, translating into enhanced ADCC against AML cells (cytotoxicity w/o TNF-α = 17.9% vs with TNF-α = 34.3%, n=13-15). Conversely, IFN-γ exposure led to reduced ADCC (cytotoxicity w/o IFN-γ = 17.9% vs with IFN-γ = 9.2%, n=15), which is attributed to increased expression of NK inhibitory receptor ligands (HLA-ABC, HLA-E). Blocking of the corresponding inhibitory NK receptors (KIR/CD158b and NKG2A) partially reversed this effect. Similar findings were observed with a CD33-directed antibody, indicating a universal resistance mechanism against ADCC-based immunotherapy in AML.</p><p><strong>Conclusions: </strong>CD70 is a promising target for NK cell-based immunotherapy in AML. However, IFN-γ-dependent upregulation of HLA molecules on AML cells contributes to resistance to ADCC. These findings underscore the need for rationale combination strategies in clinical trials to overcome this inducible immune escape mechanism.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12684112/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145677294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-04DOI: 10.1136/jitc-2025-012207
Giovanna Polcaro, Luigi Liguori, Valentina Manzo, Emanuela De Bellis, Berenice Stefanelli, Valentina Pagliara, Maddalena Langella, Alessandro Caputo, Alessandro Ottaiano, Rosanna Martinelli, Marco Cascella, Cristina Ferrone, Amelia Filippelli, Valeria Conti, Stefano Pepe, Francesco Sabbatino
<p><strong>Background: </strong>Predictive biomarkers for anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand 1 (PD-L1) therapy are needed. Here, we validated the role of PD-1 single nucleotide polymorphisms (SNPs) in predicting the development of immune-related adverse events (irAEs) in patients with advanced cancer treated with anti-PD-1/PD-L1-based immunotherapy and defined the molecular mechanisms underlying the role of identified SNP candidate.</p><p><strong>Methods: </strong>Blood samples, clinical-pathological characteristics, survival outcomes and irAEs were collected from two cohorts of patients: (1) patients with advanced cancer treated with anti-PD-1/PD-L1 alone and (2) patients with advanced non-small cell lung cancer (NSCLC) treated with anti-PD-1 in combination with platinum-based chemotherapy with or without anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) therapy. PD-1 SNPs including rs2227981, rs7421861, rs11568821, rs36084323, rs2227982 and rs10204525 were genotyped and correlated with clinical-pathological characteristics and irAEs. Putative miRNAs binding to PD-1 SNP candidate were identified by in silico analysis. Validation of miRNA binding to PD-1 SNP allele specificity as well as evaluation of the induced PD-1 modulation was performed in vitro using patient-derived peripheral blood mononuclear cells (PBMCs). Susceptibility of non-cancer cells to immune cells incubated with anti-PD-1 based on PD-1 SNP allele specificity and miRNA modulation was performed by co-culturing non-cancer human epidermal keratinocyte (HaCaT) cells and bronchial epithelial BEAS-2B cells with human leukocyte antigen (HLA)-matched PBMCs, obtained from patients with cancer treated with anti-PD-1/PD-L1-based immunotherapy and carrying a different PD-1 SNP.</p><p><strong>Results: </strong>Most of the analyzed PD-1 SNPs were not associated with the development of irAEs. In contrast, rs10204525 exhibited a significant association with the occurrence of both grade 1-2 and 3-4 irAEs in both cohorts of patients. Specifically, patients carrying C/C had a higher rate of irAEs as compared with those carrying C/T. rs10204525 mapped on 3' untranslated region (3'-UTR) region of PD-1. miR-4717-3p bound to rs10204525 based on its allele specificity. Modulation of miR-4717-3p expression as well as of miR-4717-3p binding to rs10204525 differentially regulated PD-1 expression and induction in PMBCs harboring C/C or C/T genotypes as well as their ability to recognize and destroy HLA-matched HaCaT cells, even more in the presence of anti-PD-1 therapy. Specifically, PBMCs carrying a C/T genotype displayed a significantly lower ability to recognize and destroy non-cancer cells as compared to those carrying C/C. These results were further validated by co-culturing of both BEAS-2B and HaCaT non-cancer cells with PBMCs carrying differential rs10204525 genotypes, isolated from additional patients with cancer, incubated with anti-PD-1 or anti-PD-1 in combinat
{"title":"rs10204525 binding to miR-4717-3p modulates PD-1 expression and predicts the development of immune-related adverse events in patients with advanced cancer treated with anti-PD-1/PD-L1 therapy.","authors":"Giovanna Polcaro, Luigi Liguori, Valentina Manzo, Emanuela De Bellis, Berenice Stefanelli, Valentina Pagliara, Maddalena Langella, Alessandro Caputo, Alessandro Ottaiano, Rosanna Martinelli, Marco Cascella, Cristina Ferrone, Amelia Filippelli, Valeria Conti, Stefano Pepe, Francesco Sabbatino","doi":"10.1136/jitc-2025-012207","DOIUrl":"10.1136/jitc-2025-012207","url":null,"abstract":"<p><strong>Background: </strong>Predictive biomarkers for anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand 1 (PD-L1) therapy are needed. Here, we validated the role of PD-1 single nucleotide polymorphisms (SNPs) in predicting the development of immune-related adverse events (irAEs) in patients with advanced cancer treated with anti-PD-1/PD-L1-based immunotherapy and defined the molecular mechanisms underlying the role of identified SNP candidate.</p><p><strong>Methods: </strong>Blood samples, clinical-pathological characteristics, survival outcomes and irAEs were collected from two cohorts of patients: (1) patients with advanced cancer treated with anti-PD-1/PD-L1 alone and (2) patients with advanced non-small cell lung cancer (NSCLC) treated with anti-PD-1 in combination with platinum-based chemotherapy with or without anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) therapy. PD-1 SNPs including rs2227981, rs7421861, rs11568821, rs36084323, rs2227982 and rs10204525 were genotyped and correlated with clinical-pathological characteristics and irAEs. Putative miRNAs binding to PD-1 SNP candidate were identified by in silico analysis. Validation of miRNA binding to PD-1 SNP allele specificity as well as evaluation of the induced PD-1 modulation was performed in vitro using patient-derived peripheral blood mononuclear cells (PBMCs). Susceptibility of non-cancer cells to immune cells incubated with anti-PD-1 based on PD-1 SNP allele specificity and miRNA modulation was performed by co-culturing non-cancer human epidermal keratinocyte (HaCaT) cells and bronchial epithelial BEAS-2B cells with human leukocyte antigen (HLA)-matched PBMCs, obtained from patients with cancer treated with anti-PD-1/PD-L1-based immunotherapy and carrying a different PD-1 SNP.</p><p><strong>Results: </strong>Most of the analyzed PD-1 SNPs were not associated with the development of irAEs. In contrast, rs10204525 exhibited a significant association with the occurrence of both grade 1-2 and 3-4 irAEs in both cohorts of patients. Specifically, patients carrying C/C had a higher rate of irAEs as compared with those carrying C/T. rs10204525 mapped on 3' untranslated region (3'-UTR) region of PD-1. miR-4717-3p bound to rs10204525 based on its allele specificity. Modulation of miR-4717-3p expression as well as of miR-4717-3p binding to rs10204525 differentially regulated PD-1 expression and induction in PMBCs harboring C/C or C/T genotypes as well as their ability to recognize and destroy HLA-matched HaCaT cells, even more in the presence of anti-PD-1 therapy. Specifically, PBMCs carrying a C/T genotype displayed a significantly lower ability to recognize and destroy non-cancer cells as compared to those carrying C/C. These results were further validated by co-culturing of both BEAS-2B and HaCaT non-cancer cells with PBMCs carrying differential rs10204525 genotypes, isolated from additional patients with cancer, incubated with anti-PD-1 or anti-PD-1 in combinat","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12684190/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145677267","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1136/jitc-2025-013145
Xiaonan Xiang, Fei Wang, Yanbin Shen, Sheng Dai
Common lymphatic endothelial and vascular endothelial receptor-1 (CLEVER-1) is a multifunctional scavenger receptor expressed on tumor-associated macrophages (TAMs). In a recent study published in the Journal for ImmunoTherapy of Cancer, Yu et al reported that CLEVER-1+ TAMs accumulate in advanced gastric cancer (GC), associate with poor prognosis, and contribute to resistance to chemoimmunotherapy. CLEVER-1 blockade using bexmarilimab reprogrammed TAMs toward a pro-inflammatory phenotype by suppressing peroxisome proliferator-activated receptor gamma (PPARγ)-driven lipid metabolism and enhancing antigen presentation and inflammatory cytokine secretion. CLEVER-1 blockade also synergized with anti-programmed cell death protein 1 (PD-1) therapy in ex vivo GC models, particularly in tumors enriched with CLEVER-1+ TAM. These findings identify CLEVER-1+ TAMs as both biomarker and functional mediator of anti-PD-1 therapy resistance, providing a rationale for combining bexmarilimab with immune checkpoint blockade in GC. In this commentary, we discuss the mechanistic significance, translational potential, and clinical prospects of CLEVER-1 blockade to overcome immunotherapy resistance in GC.
普通淋巴内皮和血管内皮受体-1 (clif -1)是一种表达于肿瘤相关巨噬细胞(tam)的多功能清道夫受体。最近发表在《癌症免疫治疗杂志》(Journal for ImmunoTherapy of Cancer)上的一项研究中,Yu等人报道了clev -1+ tam在晚期胃癌(GC)中蓄积,与不良预后相关,并导致对化学免疫治疗的耐药。bexmarilimab通过抑制过氧化物酶体增殖物激活受体γ (PPARγ)驱动的脂质代谢和增强抗原呈递和炎症细胞因子分泌,将TAMs重编程为促炎表型。在离体GC模型中,特别是在富含kwel -1+ TAM的肿瘤中,kwel -1阻断剂还与抗程序性细胞死亡蛋白1 (PD-1)治疗协同作用。这些发现确定了kwel -1+ tam是抗pd -1治疗耐药的生物标志物和功能介质,为bexmarilimab联合免疫检查点阻断在GC中的应用提供了理论依据。在这篇评论中,我们讨论了clai -1阻断治疗GC免疫治疗耐药的机制意义、转化潜力和临床前景。
{"title":"CLEVER-1 blockade reprograms TAMs to overcome anti-PD-1 resistance in gastric cancer.","authors":"Xiaonan Xiang, Fei Wang, Yanbin Shen, Sheng Dai","doi":"10.1136/jitc-2025-013145","DOIUrl":"10.1136/jitc-2025-013145","url":null,"abstract":"<p><p>Common lymphatic endothelial and vascular endothelial receptor-1 (CLEVER-1) is a multifunctional scavenger receptor expressed on tumor-associated macrophages (TAMs). In a recent study published in the <i>Journal for ImmunoTherapy of Cancer</i>, Yu <i>et al</i> reported that CLEVER-1<sup>+</sup> TAMs accumulate in advanced gastric cancer (GC), associate with poor prognosis, and contribute to resistance to chemoimmunotherapy. CLEVER-1 blockade using bexmarilimab reprogrammed TAMs toward a pro-inflammatory phenotype by suppressing peroxisome proliferator-activated receptor gamma (PPARγ)-driven lipid metabolism and enhancing antigen presentation and inflammatory cytokine secretion. CLEVER-1 blockade also synergized with anti-programmed cell death protein 1 (PD-1) therapy in ex vivo GC models, particularly in tumors enriched with CLEVER-1<sup>+</sup> TAM. These findings identify CLEVER-1<sup>+</sup> TAMs as both biomarker and functional mediator of anti-PD-1 therapy resistance, providing a rationale for combining bexmarilimab with immune checkpoint blockade in GC. In this commentary, we discuss the mechanistic significance, translational potential, and clinical prospects of CLEVER-1 blockade to overcome immunotherapy resistance in GC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12682175/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668298","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Blinatumomab, inotuzumab or autologous anti-CD19 chimeric antigen receptor (CAR)-T cells have revolutionized the treatment of relapsed or refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, tumor escape through antigenic modulation accounts for almost 40% of subsequent relapses. Multi-antigen targeting strategies should be developed, and it is urgent to identify new targets.
Methods: We investigated the extensive immunophenotyping of 13 BCP-ALL from pediatric patients by using the BioLegend Human Cell Surface Marker Screening Kit. Then, to assess whether targeting each antigen with monoclonal antibodies could lead to leukemic cell lysis, long-term antibody-dependent cellular cytotoxicity (ADCC) assays were performed using murine monoclonal antibodies and human T cells armed with murine CD16.
Results: 13 highly expressed antigens were selected. With the antibodies tested here, the most significant lysis was observed by targeting CD24 and CD156c. The double targeting of CD24-CD123 appeared to be even more effective. Triple targeting was associated with a reduction in ADCC activity.
Conclusion: CD24 therefore emerged as an effective target in BCP-ALL, and the combination of CD24 and CD123 as a potential effective double-targeting strategy. The combination of different recognition modalities (eg, a CAR and CD16) should be tested to determine whether it provides synergistic cytotoxic activity in triple targeting.
{"title":"Evaluation of multi-antigen targeting ADCC strategies in pediatric BCP-ALL.","authors":"Audrey Grain, Jocelyn Ollier, Baptiste Le Calvez, Elodie Guiet, Caroline Thomas, Marie-Laure Couec, Margaux Camuset, Fanny Rialland, Marion Eveillard, Emmanuel Scotet, Béatrice Clémenceau","doi":"10.1136/jitc-2025-012552","DOIUrl":"10.1136/jitc-2025-012552","url":null,"abstract":"<p><strong>Background: </strong>Blinatumomab, inotuzumab or autologous anti-CD19 chimeric antigen receptor (CAR)-T cells have revolutionized the treatment of relapsed or refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, tumor escape through antigenic modulation accounts for almost 40% of subsequent relapses. Multi-antigen targeting strategies should be developed, and it is urgent to identify new targets.</p><p><strong>Methods: </strong>We investigated the extensive immunophenotyping of 13 BCP-ALL from pediatric patients by using the BioLegend Human Cell Surface Marker Screening Kit. Then, to assess whether targeting each antigen with monoclonal antibodies could lead to leukemic cell lysis, long-term antibody-dependent cellular cytotoxicity (ADCC) assays were performed using murine monoclonal antibodies and human T cells armed with murine CD16.</p><p><strong>Results: </strong>13 highly expressed antigens were selected. With the antibodies tested here, the most significant lysis was observed by targeting CD24 and CD156c. The double targeting of CD24-CD123 appeared to be even more effective. Triple targeting was associated with a reduction in ADCC activity.</p><p><strong>Conclusion: </strong>CD24 therefore emerged as an effective target in BCP-ALL, and the combination of CD24 and CD123 as a potential effective double-targeting strategy. The combination of different recognition modalities (eg, a CAR and CD16) should be tested to determine whether it provides synergistic cytotoxic activity in triple targeting.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12682191/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1136/jitc-2025-013733
Kun Li, Lixin Niu, Xufei Zhang, Tianzi Li, Xiaoyan Zhou, Lin Wang, Jinxiang Han, Ziqiang Wang
Background: T-cell exhaustion induced by the tumor microenvironment is an important factor in posing a major challenge to effective cancer immunotherapy. Immune checkpoint inhibitors aim to reverse T-cell exhaustion. However, the effectiveness of immune checkpoint inhibitors is often limited due to their off-target effects and single targets. Herein, we attempt to identify molecular targets that can regulate the expression of multiple immune checkpoints to reverse T-cell exhaustion.
Methods: NSG mice with xenotransplantation of human bladder cancer cells were used to investigate the function of nuclear paraspeckle assembly transcript 1 (NEAT1) in T-cell exhaustion. Chromatin isolation by RNA purification, chromatin immunoprecipitation, and luciferase assays was employed to investigate the molecular mechanisms by which NEAT1 regulates expression of target genes.
Results: NEAT1, a bladder cancer-related long non-coding RNA (lncRNA), promotes lactate production in tumor cells by binding to the lactate dehydrogenase A gene. This lactate production subsequently inhibits NEAT1 expression in CD8+T cells. Furthermore, NEAT1 in CD8+T cells plays a crucial role in modulating the immune response of CD8+T cells against tumor cells. Our findings indicate that NEAT1 regulates the expression of multiple immune checkpoint genes by directly binding to them and inhibiting transcription through the alteration of histone lactylation near transcriptional start sites, which affects RNA polymerase II recruitment.
Conclusions: lncRNA NEAT1 serves as a modulator of the antitumor response of CD8+T cells in the bladder tumor microenvironment and may represent a therapeutic target for reversing T-cell exhaustion.
{"title":"NEAT1 is a therapeutic target for reversing T-cell exhaustion in bladder cancer.","authors":"Kun Li, Lixin Niu, Xufei Zhang, Tianzi Li, Xiaoyan Zhou, Lin Wang, Jinxiang Han, Ziqiang Wang","doi":"10.1136/jitc-2025-013733","DOIUrl":"10.1136/jitc-2025-013733","url":null,"abstract":"<p><strong>Background: </strong>T-cell exhaustion induced by the tumor microenvironment is an important factor in posing a major challenge to effective cancer immunotherapy. Immune checkpoint inhibitors aim to reverse T-cell exhaustion. However, the effectiveness of immune checkpoint inhibitors is often limited due to their off-target effects and single targets. Herein, we attempt to identify molecular targets that can regulate the expression of multiple immune checkpoints to reverse T-cell exhaustion.</p><p><strong>Methods: </strong>NSG mice with xenotransplantation of human bladder cancer cells were used to investigate the function of nuclear paraspeckle assembly transcript 1 (NEAT1) in T-cell exhaustion. Chromatin isolation by RNA purification, chromatin immunoprecipitation, and luciferase assays was employed to investigate the molecular mechanisms by which NEAT1 regulates expression of target genes.</p><p><strong>Results: </strong>NEAT1, a bladder cancer-related long non-coding RNA (lncRNA), promotes lactate production in tumor cells by binding to the lactate dehydrogenase A gene. This lactate production subsequently inhibits NEAT1 expression in CD8+T cells. Furthermore, NEAT1 in CD8+T cells plays a crucial role in modulating the immune response of CD8+T cells against tumor cells. Our findings indicate that NEAT1 regulates the expression of multiple immune checkpoint genes by directly binding to them and inhibiting transcription through the alteration of histone lactylation near transcriptional start sites, which affects RNA polymerase II recruitment.</p><p><strong>Conclusions: </strong>lncRNA NEAT1 serves as a modulator of the antitumor response of CD8+T cells in the bladder tumor microenvironment and may represent a therapeutic target for reversing T-cell exhaustion.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12682204/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668451","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-03DOI: 10.1136/jitc-2025-013118
Shoaib Anwaar, Amina Ashraf, Sarah Jahfali, Joseph Yunis, Jazmina L Gonzalez Cruz, James W Wells
Background: Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer in Asian, Caucasian, and Hispanic populations and its aggressive form contributes to significant morbidity and mortality. Chronic ultraviolet B (UVB) exposure is a major environmental carcinogen that drives cSCC initiation, progression, and immune evasion. Regulatory T cells (Tregs) are known mediators of UVB-induced immunosuppression; however, their direct involvement in the establishment of cSCC remains elusive.
Methods: Flow cytometry was employed to quantify Treg populations in the skin and draining lymph nodes of UVB-exposed and untreated mice. The functional role of Tregs following UVB exposure was examined using a contact hypersensitivity assay, where Treg activity was modulated by anti-cytotoxic T-lymphocyte-associated protein (CTLA)-4, anti-TIGIT, and anti-FR4 antibodies. The capacity of UVB to render mice susceptible to immunogenic cSCC tumor establishment was assessed under different UVB exposure regimens. Treg-modulating antibodies were administered following UVB treatment and prior to tumor implantation to explore whether UV-induced Treg manipulation can prevent cSCC tumor establishment.
Results: UVB irradiation for 5 consecutive days significantly increased the number of CD4+ Foxp3+ Tregs in both skin and skin-draining lymph nodes. These Tregs were shown to be suppressive in contact hypersensitivity assays. However, suppression was prevented following depletion of Tregs and/or avolition of their function using monoclonal antibodies. Consistently, chronic UVB exposure prior to tumor implantation permitted the establishment and growth of otherwise immunogenic cSCC tumors, which correlated with the expansion and recruitment of Tregs into the skin. Importantly, immunomodulation with anti-CTLA-4 or anti-FR4 after chronic UVB exposure effectively prevented cSCC establishment, indicating that the manipulation of UV-induced Tregs prevented the establishment and growth of immunogenic cSCC tumors.
Conclusion: Our findings show that the manipulation of UV-induced Tregs prevents early cSCC establishment. Thus, strategies aimed at modulating Treg function or abundance in the skin may represent a feasible therapeutic avenue for the prevention of cSCC tumor emergence in patients.
{"title":"Immunomodulation of UVB-induced regulatory T cells prevents the establishment of squamous cell carcinoma.","authors":"Shoaib Anwaar, Amina Ashraf, Sarah Jahfali, Joseph Yunis, Jazmina L Gonzalez Cruz, James W Wells","doi":"10.1136/jitc-2025-013118","DOIUrl":"10.1136/jitc-2025-013118","url":null,"abstract":"<p><strong>Background: </strong>Cutaneous squamous cell carcinoma (cSCC) is the second most common form of skin cancer in Asian, Caucasian, and Hispanic populations and its aggressive form contributes to significant morbidity and mortality. Chronic ultraviolet B (UVB) exposure is a major environmental carcinogen that drives cSCC initiation, progression, and immune evasion. Regulatory T cells (Tregs) are known mediators of UVB-induced immunosuppression; however, their direct involvement in the establishment of cSCC remains elusive.</p><p><strong>Methods: </strong>Flow cytometry was employed to quantify Treg populations in the skin and draining lymph nodes of UVB-exposed and untreated mice. The functional role of Tregs following UVB exposure was examined using a contact hypersensitivity assay, where Treg activity was modulated by anti-cytotoxic T-lymphocyte-associated protein (CTLA)-4, anti-TIGIT, and anti-FR4 antibodies. The capacity of UVB to render mice susceptible to immunogenic cSCC tumor establishment was assessed under different UVB exposure regimens. Treg-modulating antibodies were administered following UVB treatment and prior to tumor implantation to explore whether UV-induced Treg manipulation can prevent cSCC tumor establishment.</p><p><strong>Results: </strong>UVB irradiation for 5 consecutive days significantly increased the number of CD4<sup>+</sup> Foxp3<sup>+</sup> Tregs in both skin and skin-draining lymph nodes. These Tregs were shown to be suppressive in contact hypersensitivity assays. However, suppression was prevented following depletion of Tregs and/or avolition of their function using monoclonal antibodies. Consistently, chronic UVB exposure prior to tumor implantation permitted the establishment and growth of otherwise immunogenic cSCC tumors, which correlated with the expansion and recruitment of Tregs into the skin. Importantly, immunomodulation with anti-CTLA-4 or anti-FR4 after chronic UVB exposure effectively prevented cSCC establishment, indicating that the manipulation of UV-induced Tregs prevented the establishment and growth of immunogenic cSCC tumors.</p><p><strong>Conclusion: </strong>Our findings show that the manipulation of UV-induced Tregs prevents early cSCC establishment. Thus, strategies aimed at modulating Treg function or abundance in the skin may represent a feasible therapeutic avenue for the prevention of cSCC tumor emergence in patients.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12682195/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-02DOI: 10.1136/jitc-2025-013518
Mark D Stewart, Christopher R Cabanski, Jeff D Allen, John E Connolly, Ben M Beneski, Boro Dropulić, Steven A Feldman, Lee A Fleisher, Patrick J Hanley, Kristen Hege, Natasha Kekre, Holly Fernandez Lynch, Crystal L Mackall
Genetically modified cell-based therapies hold transformative potential, particularly for patients with rare cancers and ultra-rare diseases. However, progress toward regulatory approval, reimbursement, and broad patient access is often constrained by misaligned regulatory, manufacturing, and financial frameworks that do not reflect the realities of treating small populations and low-throughput production models. Drawing on a collaborative white paper and public meeting convened by Friends of Cancer Research and the Parker Institute for Cancer Immunotherapy in May 2025, this commentary outlines three strategies to streamline regulatory pathways and enable timely, sustainable access: (1) flexible approaches to Chemistry, Manufacturing, and Controls requirements in small populations, (2) adaptable regulatory frameworks to support diverse manufacturing models, and (3) limited cost recovery mechanisms to bridge early access and development gaps. Recent regulatory and policy discussions have echoed these priorities, signaling an opportunity to align oversight with operational realities to advance innovation and access for patients in high-need settings.
基于基因修饰细胞的疗法具有变革潜力,特别是对于患有罕见癌症和超罕见疾病的患者。然而,监管审批、报销和广泛的患者可及性方面的进展往往受到监管、制造和财务框架不一致的限制,这些框架没有反映治疗小人群和低通量生产模式的现实。根据癌症研究之友(Friends of Cancer Research)和帕克癌症免疫治疗研究所(Parker Institute for Cancer Immunotherapy)于2025年5月召开的合作白皮书和公开会议,本评论概述了简化监管途径并实现及时、可持续获取的三项战略:(1)在小规模人口中灵活应对化学、制造和控制需求;(2)适应性强的监管框架,以支持多样化的制造模式;(3)有限的成本回收机制,以弥合早期准入和发展差距。最近的监管和政策讨论呼应了这些优先事项,表明有机会将监督与业务现实相结合,以推进创新和高需求环境中患者的可及性。
{"title":"Enabling access to genetically modified cell therapies through flexible approaches to manufacturing and cost recovery.","authors":"Mark D Stewart, Christopher R Cabanski, Jeff D Allen, John E Connolly, Ben M Beneski, Boro Dropulić, Steven A Feldman, Lee A Fleisher, Patrick J Hanley, Kristen Hege, Natasha Kekre, Holly Fernandez Lynch, Crystal L Mackall","doi":"10.1136/jitc-2025-013518","DOIUrl":"10.1136/jitc-2025-013518","url":null,"abstract":"<p><p>Genetically modified cell-based therapies hold transformative potential, particularly for patients with rare cancers and ultra-rare diseases. However, progress toward regulatory approval, reimbursement, and broad patient access is often constrained by misaligned regulatory, manufacturing, and financial frameworks that do not reflect the realities of treating small populations and low-throughput production models. Drawing on a collaborative white paper and public meeting convened by Friends of Cancer Research and the Parker Institute for Cancer Immunotherapy in May 2025, this commentary outlines three strategies to streamline regulatory pathways and enable timely, sustainable access: (1) flexible approaches to Chemistry, Manufacturing, and Controls requirements in small populations, (2) adaptable regulatory frameworks to support diverse manufacturing models, and (3) limited cost recovery mechanisms to bridge early access and development gaps. Recent regulatory and policy discussions have echoed these priorities, signaling an opportunity to align oversight with operational realities to advance innovation and access for patients in high-need settings.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12684139/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145661256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1136/jitc-2025-013783
Tomonori Yaguchi, Kenji Chamoto, Tasuku Honjo
Aging impairs antitumor immunity and may reduce the efficacy of immune checkpoint inhibitors (ICIs). However, the underlying mechanisms remain unclear. Building on our recent findings, we review three key mechanisms of CD8+ T-cell aging: elevated T-cell receptor (TCR) activation thresholds, mitochondrial dysfunction, and disruption of proteostasis. Studies in aged mice have revealed that aged naïve T cells exhibit defective priming due to increased CD45 expression, which raises the TCR activation threshold and restricts effector differentiation. Aging also impairs mitochondrial metabolism, particularly fatty acid oxidation. Furthermore, we highlight the role of proteostasis collapse, including defective autophagy and increased endoplasmic reticulum stress, as a contributor to T-cell dysfunction. Spermidine, a polyamine that declines with age, has the potential to modulate both mitochondrial function and proteostasis. Its supplementation has been shown to improve programmed cell death-1 blockade responsiveness in aged mice. Although clinical studies in humans have yielded inconsistent results regarding the effect of chronological age on ICI efficacy, identifying patients with "age-related" immune microenvironments may enable stratified therapeutic approaches based on insights from preclinical aging models.
{"title":"Age-related immune states and PD-1 blockade: mechanisms and strategies for the elderly.","authors":"Tomonori Yaguchi, Kenji Chamoto, Tasuku Honjo","doi":"10.1136/jitc-2025-013783","DOIUrl":"10.1136/jitc-2025-013783","url":null,"abstract":"<p><p>Aging impairs antitumor immunity and may reduce the efficacy of immune checkpoint inhibitors (ICIs). However, the underlying mechanisms remain unclear. Building on our recent findings, we review three key mechanisms of CD8<sup>+</sup> T-cell aging: elevated T-cell receptor (TCR) activation thresholds, mitochondrial dysfunction, and disruption of proteostasis. Studies in aged mice have revealed that aged naïve T cells exhibit defective priming due to increased CD45 expression, which raises the TCR activation threshold and restricts effector differentiation. Aging also impairs mitochondrial metabolism, particularly fatty acid oxidation. Furthermore, we highlight the role of proteostasis collapse, including defective autophagy and increased endoplasmic reticulum stress, as a contributor to T-cell dysfunction. Spermidine, a polyamine that declines with age, has the potential to modulate both mitochondrial function and proteostasis. Its supplementation has been shown to improve programmed cell death-1 blockade responsiveness in aged mice. Although clinical studies in humans have yielded inconsistent results regarding the effect of chronological age on ICI efficacy, identifying patients with \"age-related\" immune microenvironments may enable stratified therapeutic approaches based on insights from preclinical aging models.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12673548/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145661100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1136/jitc-2025-012908
Zhuangzhuang Feng, Xinxiu Shi, Zhiyang Li, Minming Lou, Bijia Cui, Mengzhan Chen, Lei Zhang, Yujie Sun, Heng Zhang, Yi-Nan Li, Huijuan Liu, Tao Sun
Background: Enhancing immunogenicity and antigen-presentation efficiency is critical for tumor vaccine development. While yeast-surface glycoprotein side chains can improve antigen presentation, their ability to deliver tumor antigens remains limited.
Methods: The composite carbon nanopolymers (Asparagine-N-acetylglucosamine-Mannose derived carbon nanopolymers, ANM-NPs) mimicking yeast-surface glycans side chains were prepared using asparagine, N-acetylglucosamine and mannose as precursors via microwave-assisted synthesis method. Combined with whole-cell tumor antigen (T-Ag), the vaccine ANM-NPs@T-Ag was prepared. The expression levels of major histocompatibility complex-II, CD80, and CD86 were analyzed by flow cytometry to evaluate the enhancement of antigen presentation. The antitumor efficacy of ANM-NPs@T-Ag was assessed by H22-LUC and B16-F10-LUC tumor-bearing models.
Results: ANM-NPs have an average particle size of approximately 21.69 nm and exhibit good stability. Compared with traditional aluminum adjuvants, ANM-NPs demonstrate superior efficacy in promoting antigen presentation and immune activation. ANM-NPs effectively stimulated innate immunity, facilitated immune cell infiltration, and enhanced adaptive immune responses. The vaccine ANM-NPs@T-Ag targeted dendritic cells via mannose receptors, activating the lectin pathway, improving complement opsonization, and enhancing antigen presentation. ANM-NPs@T-Ag stimulated cellular/humoral immunity, increased tumor-specific IgG, and inhibited tumor growth. ANM-NPs@T-Ag also enhanced the antitumor effect of α-PD-1 (programmed cell death protein-1).
Conclusion: These findings demonstrate that ANM-NPs can serve as a potential vaccine adjuvant, and ANM-NPs@T-Ag represents a promising tumor vaccine candidate.Cite Now.
{"title":"Biomimetic carbon nanopolymers ANM-NPs act on mannose receptors and complement receptors to promote tumor antigen presentation.","authors":"Zhuangzhuang Feng, Xinxiu Shi, Zhiyang Li, Minming Lou, Bijia Cui, Mengzhan Chen, Lei Zhang, Yujie Sun, Heng Zhang, Yi-Nan Li, Huijuan Liu, Tao Sun","doi":"10.1136/jitc-2025-012908","DOIUrl":"10.1136/jitc-2025-012908","url":null,"abstract":"<p><strong>Background: </strong>Enhancing immunogenicity and antigen-presentation efficiency is critical for tumor vaccine development. While yeast-surface glycoprotein side chains can improve antigen presentation, their ability to deliver tumor antigens remains limited.</p><p><strong>Methods: </strong>The composite carbon nanopolymers (Asparagine-N-acetylglucosamine-Mannose derived carbon nanopolymers, ANM-NPs) mimicking yeast-surface glycans side chains were prepared using asparagine, N-acetylglucosamine and mannose as precursors via microwave-assisted synthesis method. Combined with whole-cell tumor antigen (T-Ag), the vaccine ANM-NPs@T-Ag was prepared. The expression levels of major histocompatibility complex-II, CD80, and CD86 were analyzed by flow cytometry to evaluate the enhancement of antigen presentation. The antitumor efficacy of ANM-NPs@T-Ag was assessed by H22-LUC and B16-F10-LUC tumor-bearing models.</p><p><strong>Results: </strong>ANM-NPs have an average particle size of approximately 21.69 nm and exhibit good stability. Compared with traditional aluminum adjuvants, ANM-NPs demonstrate superior efficacy in promoting antigen presentation and immune activation. ANM-NPs effectively stimulated innate immunity, facilitated immune cell infiltration, and enhanced adaptive immune responses. The vaccine ANM-NPs@T-Ag targeted dendritic cells via mannose receptors, activating the lectin pathway, improving complement opsonization, and enhancing antigen presentation. ANM-NPs@T-Ag stimulated cellular/humoral immunity, increased tumor-specific IgG, and inhibited tumor growth. ANM-NPs@T-Ag also enhanced the antitumor effect of α-PD-1 (programmed cell death protein-1).</p><p><strong>Conclusion: </strong>These findings demonstrate that ANM-NPs can serve as a potential vaccine adjuvant, and ANM-NPs@T-Ag represents a promising tumor vaccine candidate.Cite Now.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12673551/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145661192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1136/jitc-2025-013516
Yajing Zhang, Yu-Man Tsui, Joyce Man-Fong Lee, Tan-To Cheung, Daniel Wai-Hung Ho, Irene Oi-Lin Ng
Background: Programmed cell death protein-1 (PD-1) blockade has shown promising clinical efficacy in hepatocellular carcinoma (HCC), yet the underlying immunological mechanisms governing response and resistance remain unclear. This study aimed to delineate the temporal and spatial dynamics of T-cell clonotypes and their relationship with pathological response in neoadjuvant PD-1 blockade therapy in HCC. By integrating T-cell receptor (TCR) repertoire analysis with transcriptomic profiling, we sought to elucidate the immune landscape alterations associated with treatment outcomes.
Methods: We analyzed longitudinal, matched tumor and blood samples from our previous clinical trial (NCT05471674), encompassing 114 specimens, including pre-nivolumab tumor biopsies, post-treatment resected tumors, adjacent non-tumorous livers, and peripheral blood collected before, after, and during follow-up from 19 treatment-naïve HCC. TCR clonality, diversity, and transcriptomic signatures were assessed to characterize immune responses.
Results: Elevated clonality of intratumoral TCR clonotypes (ITCs) post-treatment correlated with increased pathological tumor necrosis, predominantly driven by the most abundant top 1% ITCs. These dominant clonotypes occupied a significantly larger clonal space and demonstrated increased spatial sharing across tumor, non-tumorous liver, and peripheral blood compartments in nivolumab responders (nivo-Rs). Shared clonotypes were positively associated with tumor necrosis extent and cytolytic activity, suggesting active immune engagement. Post-treatment, shared clonotypes exhibited peripheral expansion and greater intratumoral dominance in nivo-Rs, whereas tumor unique intratumoral TCR clonotypes (ITCs) remained prevalent in non-responders (nivo-NRs). These divergent patterns were linked to higher chemokine expression within the tumor microenvironment of nivo-Rs and impaired human leukocyte antigen (HLA) class II antigen presentation in nivo-NRs, indicating distinct immune landscape configurations influencing therapeutic response.
Conclusions: Our findings demonstrate that the peripheral infiltration and expansion of high-frequency intratumoral T-cell clonotypes are critical drivers of pathological response to neoadjuvant PD-1 blockade in HCC, highlighting potential immune biomarkers and therapeutic targets to enhance immunotherapy efficacy.
{"title":"Spatiotemporal dynamics of T cells in peripheral blood and tumor underlying differential responses to neoadjuvant PD-1 blockade in hepatocellular carcinoma.","authors":"Yajing Zhang, Yu-Man Tsui, Joyce Man-Fong Lee, Tan-To Cheung, Daniel Wai-Hung Ho, Irene Oi-Lin Ng","doi":"10.1136/jitc-2025-013516","DOIUrl":"10.1136/jitc-2025-013516","url":null,"abstract":"<p><strong>Background: </strong>Programmed cell death protein-1 (PD-1) blockade has shown promising clinical efficacy in hepatocellular carcinoma (HCC), yet the underlying immunological mechanisms governing response and resistance remain unclear. This study aimed to delineate the temporal and spatial dynamics of T-cell clonotypes and their relationship with pathological response in neoadjuvant PD-1 blockade therapy in HCC. By integrating T-cell receptor (TCR) repertoire analysis with transcriptomic profiling, we sought to elucidate the immune landscape alterations associated with treatment outcomes.</p><p><strong>Methods: </strong>We analyzed longitudinal, matched tumor and blood samples from our previous clinical trial (NCT05471674), encompassing 114 specimens, including pre-nivolumab tumor biopsies, post-treatment resected tumors, adjacent non-tumorous livers, and peripheral blood collected before, after, and during follow-up from 19 treatment-naïve HCC. TCR clonality, diversity, and transcriptomic signatures were assessed to characterize immune responses.</p><p><strong>Results: </strong>Elevated clonality of intratumoral TCR clonotypes (ITCs) post-treatment correlated with increased pathological tumor necrosis, predominantly driven by the most abundant top 1% ITCs. These dominant clonotypes occupied a significantly larger clonal space and demonstrated increased spatial sharing across tumor, non-tumorous liver, and peripheral blood compartments in nivolumab responders (nivo-Rs). Shared clonotypes were positively associated with tumor necrosis extent and cytolytic activity, suggesting active immune engagement. Post-treatment, shared clonotypes exhibited peripheral expansion and greater intratumoral dominance in nivo-Rs, whereas tumor unique intratumoral TCR clonotypes (ITCs) remained prevalent in non-responders (nivo-NRs). These divergent patterns were linked to higher chemokine expression within the tumor microenvironment of nivo-Rs and impaired human leukocyte antigen (HLA) class II antigen presentation in nivo-NRs, indicating distinct immune landscape configurations influencing therapeutic response.</p><p><strong>Conclusions: </strong>Our findings demonstrate that the peripheral infiltration and expansion of high-frequency intratumoral T-cell clonotypes are critical drivers of pathological response to neoadjuvant PD-1 blockade in HCC, highlighting potential immune biomarkers and therapeutic targets to enhance immunotherapy efficacy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12673557/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145661226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}