首页 > 最新文献

Journal for Immunotherapy of Cancer最新文献

英文 中文
Cannabidiol suppresses emergency MDSCs generation by disturbing EEF1B2-mediated C/EBPβ protein synthesis in colorectal adenomas. 大麻二酚通过干扰结直肠腺瘤中eef1b2介导的C/EBPβ蛋白合成来抑制紧急MDSCs的生成。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-03 DOI: 10.1136/jitc-2025-013081
Jie Pan, Lixin Zhao, Haojie Du, Yuyu Zhu, Xiaofan Sun, Qiang Xu, Haibo Cheng, Hongqi Chen, Yang Sun

Background: Colorectal cancer often develops from adenomas over years, necessitating early intervention. Myeloid-derived suppressor cells (MDSCs) are major immune suppressive cell types in colon cancer development from adenomas through early inflammation-induced emergency myelopoiesis. Cannabidiol (CBD) is reported to function in psychosis, coronavirus infection and some cancers through immune regulation. However, its target and underlying mechanisms in colorectal adenomas are unknown.

Methods: The antitumor effect of CBD was validated in two classical colorectal adenomas models including azoxymethane (AOM)/dextran sulfate sodium salt (DSS) induced mice model and high-fat fed Apcmin/+ mice model. Single-cell RNA sequencing was used to identified the immune environment change after CBD treatment in mice colorectal adenomas. Target responsive accessibility profiling was used to find the target of CBD in MDSCs. Subsequently, multiple immunology assays and molecular biology experiment were employed to explore the adenomas prevention mechanisms of CBD.

Results: Here, we found that CBD prevented the incidence of colorectal adenomas in AOM/DSS model and high-fat diet fed Apcmin/+ mice model. Our single-cell RNA sequencing data and the results of immunofluorescence revealed that CBD treatment significantly decreased the number of MDSCs in both two colon adenomas models. Mechanistically, CBD bound to the guanine nucleotide exchange factor domain of EEF1B2, inhibiting its function in translational elongation and subsequent C/EBPβ synthesis. This disruption suppressed the differentiation and generation of MDSCs, leading to enhanced T-cell activation and prevention of colorectal adenoma progression.

Conclusion: Our findings reveal EEF1B2-mediated C/EBPβ protein synthesis as a crucial pathway in MDSC generation and highlight the potential of CBD as an early intervention strategy for colorectal adenomas.

背景:结直肠癌通常由腺瘤发展而来,需要早期干预。髓源性抑制细胞(Myeloid-derived suppressor cells, MDSCs)是一种主要的免疫抑制细胞类型,可通过早期炎症诱导的紧急骨髓生成,从腺瘤发展为结肠癌。据报道,大麻二酚(CBD)通过免疫调节在精神病、冠状病毒感染和某些癌症中发挥作用。然而,其在结直肠腺瘤中的作用靶点和潜在机制尚不清楚。方法:采用氮氧甲烷(AOM)/硫酸葡聚糖钠盐(DSS)诱导小鼠模型和高脂喂养Apcmin/+小鼠模型验证CBD的抗肿瘤作用。采用单细胞RNA测序技术检测CBD治疗小鼠结直肠腺瘤后免疫环境的变化。目标响应性可及性分析用于寻找MDSCs中CBD的目标。随后,通过多项免疫学实验和分子生物学实验来探讨CBD预防腺瘤的机制。结果:本研究发现,在AOM/DSS模型和高脂饲料喂养Apcmin/+小鼠模型中,CBD可预防结直肠腺瘤的发生。我们的单细胞RNA测序数据和免疫荧光结果显示,CBD治疗显著减少了两种结肠腺瘤模型中MDSCs的数量。在机制上,CBD结合到EEF1B2的鸟嘌呤核苷酸交换因子结构域,抑制其在翻译延伸和随后的C/EBPβ合成中的功能。这种破坏抑制了MDSCs的分化和生成,导致t细胞活化增强,预防结直肠腺瘤的进展。结论:我们的研究结果揭示了eef1b2介导的C/EBPβ蛋白合成是MDSC生成的关键途径,并强调了CBD作为结直肠腺瘤早期干预策略的潜力。
{"title":"Cannabidiol suppresses emergency MDSCs generation by disturbing EEF1B2-mediated C/EBP<i>β</i> protein synthesis in colorectal adenomas.","authors":"Jie Pan, Lixin Zhao, Haojie Du, Yuyu Zhu, Xiaofan Sun, Qiang Xu, Haibo Cheng, Hongqi Chen, Yang Sun","doi":"10.1136/jitc-2025-013081","DOIUrl":"10.1136/jitc-2025-013081","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer often develops from adenomas over years, necessitating early intervention. Myeloid-derived suppressor cells (MDSCs) are major immune suppressive cell types in colon cancer development from adenomas through early inflammation-induced emergency myelopoiesis. Cannabidiol (CBD) is reported to function in psychosis, coronavirus infection and some cancers through immune regulation. However, its target and underlying mechanisms in colorectal adenomas are unknown.</p><p><strong>Methods: </strong>The antitumor effect of CBD was validated in two classical colorectal adenomas models including azoxymethane (AOM)/dextran sulfate sodium salt (DSS) induced mice model and high-fat fed <i>Apc<sup>min/+</sup></i> mice model. Single-cell RNA sequencing was used to identified the immune environment change after CBD treatment in mice colorectal adenomas. Target responsive accessibility profiling was used to find the target of CBD in MDSCs. Subsequently, multiple immunology assays and molecular biology experiment were employed to explore the adenomas prevention mechanisms of CBD.</p><p><strong>Results: </strong>Here, we found that CBD prevented the incidence of colorectal adenomas in AOM/DSS model and high-fat diet fed <i>Apc<sup>min/+</sup></i> mice model. Our single-cell RNA sequencing data and the results of immunofluorescence revealed that CBD treatment significantly decreased the number of MDSCs in both two colon adenomas models. Mechanistically, CBD bound to the guanine nucleotide exchange factor domain of EEF1B2, inhibiting its function in translational elongation and subsequent C/EBP<i>β</i> synthesis. This disruption suppressed the differentiation and generation of MDSCs, leading to enhanced T-cell activation and prevention of colorectal adenoma progression.</p><p><strong>Conclusion: </strong>Our findings reveal EEF1B2-mediated C/EBP<i>β</i> protein synthesis as a crucial pathway in MDSC generation and highlight the potential of CBD as an early intervention strategy for colorectal adenomas.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766802/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145900379","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}
引用次数: 0
Mechanistic insights into PD-1 inhibitor-associated immune thrombocytopenia: a case report integrating longitudinal transcriptomic profiling. 机制洞察PD-1抑制剂相关的免疫性血小板减少症:一个案例报告整合纵向转录组分析。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-31 DOI: 10.1136/jitc-2025-013108
Huishan Li, Yan Wang, Jianfeng Zhu, Hehui Chen, Tianshu Liu, Luoyan Ai

Immune checkpoint inhibitors (ICIs) have significantly improved survival outcomes in patients with advanced malignancies. However, their association with immune thrombocytopenia (ITP) poses a critical risk of severe hemorrhage and necessitates treatment discontinuation. The precise molecular mechanisms underlying ICI-associated ITP remain largely unclear, critically impeding the development of predictive biomarkers and targeted therapeutic strategies. Here, we present a case report of an elderly patient with stage IV gastric adenocarcinoma who developed recurrent ITP following programmed death receptor-1 (PD-1) inhibitor serplulimab, trastuzumab and chemotherapy. Notably, longitudinal peripheral blood mononuclear cell samples were collected and subjected to transcriptomic profiling before and after two distinct ITP episodes: the initial occurrence and a recurrence triggered by PD-1 inhibitor rechallenge, providing matched time-resolved data for mechanistic analysis. Our findings demonstrate that ICIs-induced ITP involves a dual pathogenic mechanism combining immune-mediated platelet destruction and intrinsic megakaryopoietic impairment providing a novel conceptual framework for understanding this immunotherapy complication. And this represents the first prospective longitudinal investigation combining serial biospecimen collection with transcriptomic profiling (RNA sequencing) to elucidate mechanisms underlying ICIs-induced ITP.

免疫检查点抑制剂(ICIs)显著改善了晚期恶性肿瘤患者的生存结果。然而,它们与免疫性血小板减少症(ITP)的关联导致严重出血的危险,需要停止治疗。ici相关ITP的精确分子机制在很大程度上仍不清楚,严重阻碍了预测性生物标志物和靶向治疗策略的发展。在这里,我们报告了一例老年IV期胃腺癌患者,在应用程序性死亡受体-1 (PD-1)抑制剂serpluliumab、曲妥珠单抗和化疗后发生复发性ITP。值得注意的是,收集纵向外周血单个核细胞样本,并在两次不同的ITP发作之前和之后进行转录组学分析:首次发生和由PD-1抑制剂再挑战引发的复发,为机制分析提供了匹配的时间分辨率数据。我们的研究结果表明,icis诱导的ITP涉及免疫介导的血小板破坏和内在巨核生成损伤的双重致病机制,为理解这种免疫治疗并发症提供了新的概念框架。这是首次将生物标本收集与转录组学分析(RNA测序)相结合的前瞻性纵向研究,旨在阐明icis诱导ITP的机制。
{"title":"Mechanistic insights into PD-1 inhibitor-associated immune thrombocytopenia: a case report integrating longitudinal transcriptomic profiling.","authors":"Huishan Li, Yan Wang, Jianfeng Zhu, Hehui Chen, Tianshu Liu, Luoyan Ai","doi":"10.1136/jitc-2025-013108","DOIUrl":"10.1136/jitc-2025-013108","url":null,"abstract":"<p><p>Immune checkpoint inhibitors (ICIs) have significantly improved survival outcomes in patients with advanced malignancies. However, their association with immune thrombocytopenia (ITP) poses a critical risk of severe hemorrhage and necessitates treatment discontinuation. The precise molecular mechanisms underlying ICI-associated ITP remain largely unclear, critically impeding the development of predictive biomarkers and targeted therapeutic strategies. Here, we present a case report of an elderly patient with stage IV gastric adenocarcinoma who developed recurrent ITP following programmed death receptor-1 (PD-1) inhibitor serplulimab, trastuzumab and chemotherapy. Notably, longitudinal peripheral blood mononuclear cell samples were collected and subjected to transcriptomic profiling before and after two distinct ITP episodes: the initial occurrence and a recurrence triggered by PD-1 inhibitor rechallenge, providing matched time-resolved data for mechanistic analysis. Our findings demonstrate that ICIs-induced ITP involves a dual pathogenic mechanism combining immune-mediated platelet destruction and intrinsic megakaryopoietic impairment providing a novel conceptual framework for understanding this immunotherapy complication. And this represents the first prospective longitudinal investigation combining serial biospecimen collection with transcriptomic profiling (RNA sequencing) to elucidate mechanisms underlying ICIs-induced ITP.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766837/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878314","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}
引用次数: 0
Implantable artificial lymph node enables rapid in vivo neoantigen-specific T-cell generation and expansion for improving antitumor immunity. 植入式人工淋巴结使体内新抗原特异性t细胞快速生成和扩增,以提高抗肿瘤免疫。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-31 DOI: 10.1136/jitc-2025-013043
Zhixiong Cai, Kongying Lin, Xiuqing Dong, Zhenli Li, Geng Chen, Luobin Guo, Pan Mou, Peizhe Chen, Junjing Huang, Ling Li, Xiaolong Liu, Ruijing Tang, Yongyi Zeng

Background: Tumor neoantigen vaccines typically require multiple prime-boost immunizations over several weeks or even months to elicit sufficient neoantigen-specific T-cell responses, which greatly limits their effectiveness against rapidly progressing cancers, such as hepatocellular carcinoma.

Methods: Here, we developed a functionalized alginate scaffold for T-cell activation as an artificial lymph node (FAST-LN) to rapidly and efficiently generate tumor neoantigen-specific T cells in vivo. The FAST-LN consists of an alginate scaffold crosslinked with calcium ions and hyaluronic acids conjugated with anti-TIM-3 (T cell immunoglobulin and mucin domain-containing protein-3) antibodies, then incorporates tumor neoantigen-encoding adenoviral vaccines, dendritic cells (DCs), and various cytokines (C-C motif chemokine ligand 21 (CCL21), interleukin (IL)-2, IL-4, and Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF)) that maintain DC maturation and recruit T cells for antigen presentation.

Results: This design facilitates efficient DC-T cell interactions within FAST-LN, enabling rapid generation of neoantigen-specific T cells for antitumor responses. In vivo antitumor studies demonstrated superior therapeutic efficacy of FAST-LN over traditional vaccines in mouse tumor models. Mechanistically, FAST-LN significantly enhanced tumor infiltration of CD8+ T cells, Th1 cells and macrophages, orchestrating robust antitumor responses.

Conclusions: Our results demonstrate that implantable FAST-LN rapidly induces tumor neoantigen-specific T cells in vivo, offering a promising strategy for neoantigen-based cancer immunotherapy.

背景:肿瘤新抗原疫苗通常需要在几周甚至几个月的时间内多次免疫,以引起足够的新抗原特异性t细胞反应,这极大地限制了它们对快速进展的癌症(如肝细胞癌)的有效性。方法:在此,我们开发了一种功能化海藻酸盐支架,用于T细胞活化作为人工淋巴结(FAST-LN),以快速有效地在体内产生肿瘤新抗原特异性T细胞。FAST-LN由藻酸盐支架与钙离子交联的透明质酸与anti-TIM-3共轭(T细胞免疫球蛋白和粘蛋白domain-containing蛋白质3)抗体,然后合并肿瘤neoantigen-encoding adenoviral疫苗,树突状细胞(dc)和各种细胞因子(主题趋化因子配体21 (CCL21)、白介素(IL) 2、IL - 4,粒细胞-巨噬细胞集落刺激因子(GM-CSF))维持DC成熟并募集T细胞供抗原呈递。结果:这种设计促进了FAST-LN内DC-T细胞的有效相互作用,使新抗原特异性T细胞能够快速产生抗肿瘤反应。体内抗肿瘤研究表明FAST-LN在小鼠肿瘤模型中的治疗效果优于传统疫苗。在机制上,FAST-LN显著增强CD8+ T细胞、Th1细胞和巨噬细胞的肿瘤浸润,协调强大的抗肿瘤反应。结论:我们的研究结果表明,植入式FAST-LN在体内快速诱导肿瘤新抗原特异性T细胞,为基于新抗原的癌症免疫治疗提供了一种有前景的策略。
{"title":"Implantable artificial lymph node enables rapid in vivo neoantigen-specific T-cell generation and expansion for improving antitumor immunity.","authors":"Zhixiong Cai, Kongying Lin, Xiuqing Dong, Zhenli Li, Geng Chen, Luobin Guo, Pan Mou, Peizhe Chen, Junjing Huang, Ling Li, Xiaolong Liu, Ruijing Tang, Yongyi Zeng","doi":"10.1136/jitc-2025-013043","DOIUrl":"10.1136/jitc-2025-013043","url":null,"abstract":"<p><strong>Background: </strong>Tumor neoantigen vaccines typically require multiple prime-boost immunizations over several weeks or even months to elicit sufficient neoantigen-specific T-cell responses, which greatly limits their effectiveness against rapidly progressing cancers, such as hepatocellular carcinoma.</p><p><strong>Methods: </strong>Here, we developed a functionalized alginate scaffold for T-cell activation as an artificial lymph node (FAST-LN) to rapidly and efficiently generate tumor neoantigen-specific T cells in vivo. The FAST-LN consists of an alginate scaffold crosslinked with calcium ions and hyaluronic acids conjugated with anti-TIM-3 (T cell immunoglobulin and mucin domain-containing protein-3) antibodies, then incorporates tumor neoantigen-encoding adenoviral vaccines, dendritic cells (DCs), and various cytokines (C-C motif chemokine ligand 21 (CCL21), interleukin (IL)-2, IL-4, and Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF)) that maintain DC maturation and recruit T cells for antigen presentation.</p><p><strong>Results: </strong>This design facilitates efficient DC-T cell interactions within FAST-LN, enabling rapid generation of neoantigen-specific T cells for antitumor responses. In vivo antitumor studies demonstrated superior therapeutic efficacy of FAST-LN over traditional vaccines in mouse tumor models. Mechanistically, FAST-LN significantly enhanced tumor infiltration of CD8<sup>+</sup> T cells, Th1 cells and macrophages, orchestrating robust antitumor responses.</p><p><strong>Conclusions: </strong>Our results demonstrate that implantable FAST-LN rapidly induces tumor neoantigen-specific T cells in vivo, offering a promising strategy for neoantigen-based cancer immunotherapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766791/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878346","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}
引用次数: 0
Single-cell and spatial transcriptome profiling identifies the immunosuppressive spatial niche in KRAS-mutant colorectal cancer. 单细胞和空间转录组分析鉴定kras突变型结直肠癌的免疫抑制空间生态位。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-31 DOI: 10.1136/jitc-2025-013763
Sheng Yang, Chao Gu, Xinsheng Miao, Hao Zuo, Wei Xu, Yan Zhang, Wei Tang, Jianhua Zhu, Zheng Yuan, Xinhua Gu, Chenyi Zhong, Yueming Sun, Jiahui Zhou

Background: KRAS is one of the most frequently mutated genes in colorectal cancer (CRC) and plays a crucial role in tumorigenesis, progression, immune evasion, and treatment resistance. The pronounced heterogeneity within KRAS-mutant CRC highlights the urgent need for more precise and personalized therapeutic approaches.

Methods: To investigate this heterogeneity, we employed single-cell RNA sequencing and spatial transcriptomics to comprehensively characterize the tumor microenvironment of KRAS-mutant CRC. Data preprocessing and clustering were performed using Scanpy. Spatial cell-type deconvolution was conducted via Cell2location, whereas intercellular communication and spatial dependencies were analyzed using CellChat, MISTy, and stLearn.

Results: Our analyses revealed that KRAS-mutant tumor epithelial cells recruit Mono_S100A8 monocytes via the MDK_SDC4 signaling axis. Concurrently, surrounding Fib_CTHRC1 fibroblasts secrete collagen, which interacts with integrin receptors on KRAS-mutant epithelial cells and contributes to the exclusion of lymphocyte infiltration.

Conclusion: These cellular components collaboratively established an immunosuppressive spatial niche. These findings offer novel theoretical insights and potential targets for the development of immunoregulatory strategies tailored to KRAS-mutant CRC.

背景:KRAS是结直肠癌(CRC)中最常见的突变基因之一,在肿瘤发生、进展、免疫逃避和治疗耐药中起着至关重要的作用。kras突变型结直肠癌的明显异质性突出了迫切需要更精确和个性化的治疗方法。方法:为了研究这种异质性,我们采用单细胞RNA测序和空间转录组学来全面表征kras突变型CRC的肿瘤微环境。使用Scanpy进行数据预处理和聚类。通过Cell2location进行空间细胞型反卷积,而使用CellChat、MISTy和stLearn分析细胞间通信和空间依赖性。结果:我们的分析显示,kras突变的肿瘤上皮细胞通过MDK_SDC4信号轴募集Mono_S100A8单核细胞。同时,周围的Fib_CTHRC1成纤维细胞分泌胶原,胶原与kras突变上皮细胞上的整合素受体相互作用,有助于排除淋巴细胞浸润。结论:这些细胞成分共同建立了免疫抑制空间生态位。这些发现为开发针对kras突变型CRC的免疫调节策略提供了新的理论见解和潜在靶点。
{"title":"Single-cell and spatial transcriptome profiling identifies the immunosuppressive spatial niche in <i>KRAS</i>-mutant colorectal cancer.","authors":"Sheng Yang, Chao Gu, Xinsheng Miao, Hao Zuo, Wei Xu, Yan Zhang, Wei Tang, Jianhua Zhu, Zheng Yuan, Xinhua Gu, Chenyi Zhong, Yueming Sun, Jiahui Zhou","doi":"10.1136/jitc-2025-013763","DOIUrl":"10.1136/jitc-2025-013763","url":null,"abstract":"<p><strong>Background: </strong><i>KRAS</i> is one of the most frequently mutated genes in colorectal cancer (CRC) and plays a crucial role in tumorigenesis, progression, immune evasion, and treatment resistance. The pronounced heterogeneity within <i>KRAS</i>-mutant CRC highlights the urgent need for more precise and personalized therapeutic approaches.</p><p><strong>Methods: </strong>To investigate this heterogeneity, we employed single-cell RNA sequencing and spatial transcriptomics to comprehensively characterize the tumor microenvironment of <i>KRAS</i>-mutant CRC. Data preprocessing and clustering were performed using Scanpy. Spatial cell-type deconvolution was conducted via Cell2location, whereas intercellular communication and spatial dependencies were analyzed using CellChat, MISTy, and stLearn.</p><p><strong>Results: </strong>Our analyses revealed that <i>KRAS</i>-mutant tumor epithelial cells recruit Mono_<i>S100A8</i> monocytes via the MDK_SDC4 signaling axis. Concurrently, surrounding Fib_<i>CTHRC1</i> fibroblasts secrete collagen, which interacts with integrin receptors on <i>KRAS</i>-mutant epithelial cells and contributes to the exclusion of lymphocyte infiltration.</p><p><strong>Conclusion: </strong>These cellular components collaboratively established an immunosuppressive spatial niche. These findings offer novel theoretical insights and potential targets for the development of immunoregulatory strategies tailored to <i>KRAS</i>-mutant CRC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766835/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878376","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}
引用次数: 0
Chimeric antigen receptor dendritic cells suppress melanoma growth in preclinical cancer models. 嵌合抗原受体树突状细胞在临床前癌症模型中抑制黑色素瘤生长。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-31 DOI: 10.1136/jitc-2025-013040
Julia Minnee, Wallace Zh Wong, Benjamin S Russell, Jose Lm Semana, Ruofan Li, Nathaniel R Landau, Takuya Tada

Background: Chimeric antigen receptor (CAR)-T cell therapy has been successful for the treatment of hematological cancers but less effective against solid tumors, a phenomenon that results from the immunosuppressive nature of the tumor microenvironment. As a strategy to improve the treatment of solid tumors, we applied CAR therapy to dendritic cells (DCs) to generate CAR-DCs. The CAR targeted the human epidermal growth factor receptor 2 (HER2) which is overexpressed in breast cancer to defeat the immunosuppressive nature of the tumor microenvironment.

Methods: CAR-DCs were generated by lentiviral vector transduction of SAMHD1 knock-out murine bone marrow-derived DCs. The vectors coexpressed CD40L and a soluble form of programmed cell death 1 (PD-1), a checkpoint inhibitor. To increase the durability of CAR-DCs, a gene encoding the cytokine GM-CSF was introduced into the CAR vector. The CAR-DCs were injected into mice bearing B16.HER2 melanoma tumors. Tumor growth was measured, and T cell functionality was determined by IFNγ expression and in vitro cytolytic assay.

Results: CAR-DCs suppressed the growth of B16.HER2 tumors and induced the proliferation and activation of tumor-infiltrating cytolytic CD8+T cells. The PD-1 checkpoint inhibitor further augmented the antitumor response and prevented T cell exhaustion. Vectored expression of GM-CSF increased the durability of the antitumor response.

Conclusions: CAR-DCs could be an effective strategy for therapies against solid tumors that should be further explored. The approach relies on the antigen-presenting ability of DCs and their role in T cell activation and can be coupled with checkpoint inhibition in place of monoclonal antibody treatment.

背景:嵌合抗原受体(CAR)-T细胞疗法已成功治疗血液病,但对实体瘤效果较差,这一现象是由肿瘤微环境的免疫抑制性质造成的。作为一种改善实体肿瘤治疗的策略,我们将CAR疗法应用于树突状细胞(dc)以生成CAR- dc。这种CAR靶向在乳腺癌中过度表达的人表皮生长因子受体2 (HER2),以击败肿瘤微环境的免疫抑制性质。方法:采用慢病毒载体转导SAMHD1敲除小鼠骨髓源性dc生成car - dc。这些载体共表达CD40L和一种可溶形式的程序性细胞死亡1 (PD-1),一种检查点抑制剂。为了增加CAR- dc的持久性,将编码细胞因子GM-CSF的基因引入CAR载体。将car - dc注射到B16小鼠体内。HER2黑色素瘤。测量肿瘤生长,并通过IFNγ表达和体外细胞溶解试验测定T细胞功能。结果:CAR-DCs抑制B16的生长。并诱导肿瘤浸润性细胞溶解性CD8+T细胞的增殖和活化。PD-1检查点抑制剂进一步增强了抗肿瘤反应并阻止T细胞衰竭。GM-CSF的载体表达增加了抗肿瘤反应的持久性。结论:car - dc可能是一种治疗实体瘤的有效策略,值得进一步探索。该方法依赖于dc的抗原呈递能力及其在T细胞活化中的作用,并且可以与检查点抑制结合以代替单克隆抗体治疗。
{"title":"Chimeric antigen receptor dendritic cells suppress melanoma growth in preclinical cancer models.","authors":"Julia Minnee, Wallace Zh Wong, Benjamin S Russell, Jose Lm Semana, Ruofan Li, Nathaniel R Landau, Takuya Tada","doi":"10.1136/jitc-2025-013040","DOIUrl":"https://doi.org/10.1136/jitc-2025-013040","url":null,"abstract":"<p><strong>Background: </strong>Chimeric antigen receptor (CAR)-T cell therapy has been successful for the treatment of hematological cancers but less effective against solid tumors, a phenomenon that results from the immunosuppressive nature of the tumor microenvironment. As a strategy to improve the treatment of solid tumors, we applied CAR therapy to dendritic cells (DCs) to generate CAR-DCs. The CAR targeted the human epidermal growth factor receptor 2 (HER2) which is overexpressed in breast cancer to defeat the immunosuppressive nature of the tumor microenvironment.</p><p><strong>Methods: </strong>CAR-DCs were generated by lentiviral vector transduction of SAMHD1 knock-out murine bone marrow-derived DCs. The vectors coexpressed CD40L and a soluble form of programmed cell death 1 (PD-1), a checkpoint inhibitor. To increase the durability of CAR-DCs, a gene encoding the cytokine GM-CSF was introduced into the CAR vector. The CAR-DCs were injected into mice bearing B16.HER2 melanoma tumors. Tumor growth was measured, and T cell functionality was determined by IFNγ expression and in vitro cytolytic assay.</p><p><strong>Results: </strong>CAR-DCs suppressed the growth of B16.HER2 tumors and induced the proliferation and activation of tumor-infiltrating cytolytic CD8+T cells. The PD-1 checkpoint inhibitor further augmented the antitumor response and prevented T cell exhaustion. Vectored expression of GM-CSF increased the durability of the antitumor response.</p><p><strong>Conclusions: </strong>CAR-DCs could be an effective strategy for therapies against solid tumors that should be further explored. The approach relies on the antigen-presenting ability of DCs and their role in T cell activation and can be coupled with checkpoint inhibition in place of monoclonal antibody treatment.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145878324","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Clinical-radiomics nomogram integrating lymph node radiomic features to predict immunotherapy response in advanced biliary tract cancers. 结合淋巴结放射学特征预测晚期胆道肿瘤免疫治疗反应的临床放射组学nomogram。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-30 DOI: 10.1136/jitc-2025-012416
Qian Li, Jiamin Guo, Fang Wang, Haopeng Yu, Yi Wei, Yichen Zheng, Zixing Huang, Ji Ma

Background: Immunotherapy is considered a promising treatment approach for advanced biliary tract cancers (BTCs), but only a small number of patients can respond to immunotherapy. This study aimed to develop and validate a clinical-radiomics nomogram integrating radiomic features from lymph nodes (LNs) for predicting immunotherapy efficacy in advanced BTCs.

Methods: A total of 258 patients with advanced BTCs were enrolled, comprising 206 patients in the retrospective cohort and 52 patients in the prospective cohort. Radiomic features were extracted from the LNs. The maximum relevance and minimum redundancy and least absolute shrinkage and selection operator were used to develop the radiomics signature (Rad-score). Univariate analysis and multivariate logistic regression (LR) were used to construct the clinical model. A clinical-radiomics nomogram was constructed using LR. The performance of all the models was analyzed using receiver operating characteristic curves.

Results: Nine radiomic features were combined to construct the Rad-score. The nomogram incorporated the six clinical parameters and the Rad-score, and achieved the best discriminative ability with the areas under the curve (AUCs) of 0.899, 0.843 and 0.874 in the training, validation and testing cohorts. The clinical model showed better predictive performance than the Rad-score with the AUCs of 0.834, 0.878 and 0.740 in the training, validation and testing cohorts. The calibration curve and Brier score indicated the goodness-of-fit of the nomogram. Patients with higher nomogram scores had better overall survival (OS) and progression-free survival (PFS) in comparison to those with low scores.

Conclusion: The clinical-radiomics nomogram showed promising performance for predicting the response to immunotherapy in patients with advanced BTCs.

背景:免疫治疗被认为是晚期胆道肿瘤(btc)的一种有前景的治疗方法,但只有少数患者对免疫治疗有反应。本研究旨在开发和验证结合淋巴结放射学特征的临床放射组学图,以预测晚期btc的免疫治疗效果。方法:共纳入258例晚期btc患者,其中回顾性队列206例,前瞻性队列52例。从LNs中提取放射性特征。使用最大相关性、最小冗余和最小绝对收缩和选择算子来开发放射组学签名(Rad-score)。采用单因素分析和多因素logistic回归(LR)构建临床模型。应用LR构建临床放射组学图。利用接收机工作特性曲线对各模型的性能进行了分析。结果:将9个放射学特征结合起来构建rad评分。该nomogram综合了6个临床参数和Rad-score,训练组、验证组和测试组的曲线下面积(auc)分别为0.899、0.843和0.874,其判别能力最好。训练组、验证组和测试组的auc分别为0.834、0.878和0.740,临床模型的预测效果优于rad评分。校正曲线和Brier评分表明nomogram的拟合优度。nomogram评分较高的患者比评分较低的患者有更好的总生存期(OS)和无进展生存期(PFS)。结论:临床放射组学图在预测晚期btc患者免疫治疗反应方面表现良好。
{"title":"Clinical-radiomics nomogram integrating lymph node radiomic features to predict immunotherapy response in advanced biliary tract cancers.","authors":"Qian Li, Jiamin Guo, Fang Wang, Haopeng Yu, Yi Wei, Yichen Zheng, Zixing Huang, Ji Ma","doi":"10.1136/jitc-2025-012416","DOIUrl":"10.1136/jitc-2025-012416","url":null,"abstract":"<p><strong>Background: </strong>Immunotherapy is considered a promising treatment approach for advanced biliary tract cancers (BTCs), but only a small number of patients can respond to immunotherapy. This study aimed to develop and validate a clinical-radiomics nomogram integrating radiomic features from lymph nodes (LNs) for predicting immunotherapy efficacy in advanced BTCs.</p><p><strong>Methods: </strong>A total of 258 patients with advanced BTCs were enrolled, comprising 206 patients in the retrospective cohort and 52 patients in the prospective cohort. Radiomic features were extracted from the LNs. The maximum relevance and minimum redundancy and least absolute shrinkage and selection operator were used to develop the radiomics signature (Rad-score). Univariate analysis and multivariate logistic regression (LR) were used to construct the clinical model. A clinical-radiomics nomogram was constructed using LR. The performance of all the models was analyzed using receiver operating characteristic curves.</p><p><strong>Results: </strong>Nine radiomic features were combined to construct the Rad-score. The nomogram incorporated the six clinical parameters and the Rad-score, and achieved the best discriminative ability with the areas under the curve (AUCs) of 0.899, 0.843 and 0.874 in the training, validation and testing cohorts. The clinical model showed better predictive performance than the Rad-score with the AUCs of 0.834, 0.878 and 0.740 in the training, validation and testing cohorts. The calibration curve and Brier score indicated the goodness-of-fit of the nomogram. Patients with higher nomogram scores had better overall survival (OS) and progression-free survival (PFS) in comparison to those with low scores.</p><p><strong>Conclusion: </strong>The clinical-radiomics nomogram showed promising performance for predicting the response to immunotherapy in patients with advanced BTCs.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766834/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862845","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}
引用次数: 0
MAVS/CMTM6 axis couples mitochondrial homeostasis to immunogenic senescence via CCL3-driven T-cell recruitment in renal carcinoma. 肾癌中MAVS/CMTM6轴通过ccl3驱动的t细胞募集将线粒体稳态与免疫原性衰老耦联。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-30 DOI: 10.1136/jitc-2025-011477
Hanfeng Wang, Yang Fan, Qiyang Liang, Wen Tao, Xinran Chen, Jichen Wang, Senming Cao, Jiali Ye, Shidong Zuo, Chi Zhang, Donglai Shen, Yu Gao, Qingbo Huang, Xin Ma, Xu Zhang, Yan Huang, Minghui Yang

Background: Mitochondrial antiviral signaling protein (MAVS), a central adaptor in cytosolic RNA sensing, is critical for antitumor innate immunity and maintains mitochondrial homeostasis via its mitochondrial localization. Mitochondrial dysfunction acts as a key driver and amplifier of the senescence-associated secretory phenotype (SASP), a double-edged sword in tumor progression. However, whether tumor-intrinsic MAVS can regulate antitumor immunity via cellular senescence independently of its well-established interferon signaling remains unclear.

Methods: Our study employed an integrated strategy. Clinically, we profiled MAVS expression and its association with prognosis and immune infiltration in renal tumor specimens. Mechanistic insights into tumor-intrinsic MAVS were gained through a battery of techniques spanning quantitative PCR, immunoblotting, RNA sequencing, senescence and mitochondrial function assays, confocal imaging, immunohistochemical, mass spectrometry, and co-immunoprecipitation. In vivo, we used MAVS-deficient models combined with CD8+ T-cell depletion, programmed cell death protein-1 (PD-1) blockade, or reactive oxygen species (ROS) scavenging by N-acetylcysteine (NAC), with immune infiltration characterized by flow cytometry.

Results: Clinical evidence links elevated MAVS expression in renal tumors to poor prognosis and diminished CD8+ T-cell infiltration. Strikingly, tumor-intrinsic MAVS deficiency curbed malignant progression by triggering cellular senescence and fostering a permissive niche for CD8+ T-cell activation and recruitment. Mechanistically, MAVS orchestrates mitochondrial integrity by co-localizing with and stabilizing chemokine-like factor-like MARVEL transmembrane domain-containing 6 (CMTM6), thereby shielding it from lysosomal degradation. Disruption of this axis provoked mitochondrial dysfunction and ROS accumulation, culminating in senescence and an SASP marked by chemokine C-C motif ligand 3 (CCL3). Thus, despite dampening canonical innate immune signaling, MAVS deletion unleashed potent antitumor immunity via CCL3-mediated CD8+ T-cell recruitment, an effect abolished by CD8+ T-cell depletion or ROS scavenging with NAC. Leveraging this paradigm, we demonstrated that tumor-specific MAVS deficiency acts synergistically with PD-1 blockade to achieve robust therapeutic efficacy.

Conclusions: Our findings establish the tumor-intrinsic MAVS/CMTM6/CCL3 axis as a previously unrecognized critical regulator of senescence-driven antitumor immunity in renal carcinoma. Therapeutic targeting of this axis presents a promising strategy to curtail tumor progression and potentiate immunotherapy.

背景:线粒体抗病毒信号蛋白(MAVS)是细胞质RNA传感的中心适配体,在抗肿瘤先天免疫和通过线粒体定位维持线粒体稳态中起着关键作用。线粒体功能障碍是衰老相关分泌表型(SASP)的关键驱动因素和放大器,是肿瘤进展中的一把双刃剑。然而,肿瘤固有的MAVS是否可以独立于其已建立的干扰素信号,通过细胞衰老调节抗肿瘤免疫仍不清楚。方法:本研究采用综合策略。在临床上,我们分析了MAVS的表达及其与肾肿瘤标本预后和免疫浸润的关系。通过一系列技术,包括定量PCR、免疫印迹、RNA测序、衰老和线粒体功能测定、共聚焦成像、免疫组织化学、质谱和共免疫沉淀,我们获得了肿瘤固有MAVS的机制见解。在体内,我们使用mavs缺陷模型联合CD8+ t细胞衰竭,程序性细胞死亡蛋白-1 (PD-1)阻断,或n-乙酰半胱氨酸(NAC)清除活性氧(ROS),流式细胞术表征免疫浸润。结果:临床证据表明,肾脏肿瘤中MAVS表达升高与预后不良和CD8+ t细胞浸润减少有关。引人注目的是,肿瘤固有的MAVS缺陷通过触发细胞衰老和培养CD8+ t细胞激活和募集的容位来抑制恶性进展。在机制上,MAVS通过与趋化因子样漫威跨膜结构域6 (CMTM6)共定位和稳定来协调线粒体完整性,从而保护其免受溶酶体降解。这条轴的破坏引起线粒体功能障碍和ROS积累,最终导致衰老和以趋化因子C-C基序配体3 (CCL3)为标志的SASP。因此,尽管抑制了典型的先天免疫信号,MAVS缺失通过ccl3介导的CD8+ t细胞募集释放出有效的抗肿瘤免疫,这种效应被CD8+ t细胞耗尽或NAC清除ROS所消除。利用这种模式,我们证明了肿瘤特异性MAVS缺陷与PD-1阻断协同作用,以实现强大的治疗效果。结论:我们的研究结果表明,肿瘤固有的MAVS/CMTM6/CCL3轴是肾癌衰老驱动的抗肿瘤免疫的一个先前未被认识的关键调节因子。该轴的治疗靶向提出了一种有希望的策略来抑制肿瘤进展和增强免疫治疗。
{"title":"MAVS/CMTM6 axis couples mitochondrial homeostasis to immunogenic senescence via CCL3-driven T-cell recruitment in renal carcinoma.","authors":"Hanfeng Wang, Yang Fan, Qiyang Liang, Wen Tao, Xinran Chen, Jichen Wang, Senming Cao, Jiali Ye, Shidong Zuo, Chi Zhang, Donglai Shen, Yu Gao, Qingbo Huang, Xin Ma, Xu Zhang, Yan Huang, Minghui Yang","doi":"10.1136/jitc-2025-011477","DOIUrl":"10.1136/jitc-2025-011477","url":null,"abstract":"<p><strong>Background: </strong>Mitochondrial antiviral signaling protein (MAVS), a central adaptor in cytosolic RNA sensing, is critical for antitumor innate immunity and maintains mitochondrial homeostasis via its mitochondrial localization. Mitochondrial dysfunction acts as a key driver and amplifier of the senescence-associated secretory phenotype (SASP), a double-edged sword in tumor progression. However, whether tumor-intrinsic MAVS can regulate antitumor immunity via cellular senescence independently of its well-established interferon signaling remains unclear.</p><p><strong>Methods: </strong>Our study employed an integrated strategy. Clinically, we profiled MAVS expression and its association with prognosis and immune infiltration in renal tumor specimens. Mechanistic insights into tumor-intrinsic MAVS were gained through a battery of techniques spanning quantitative PCR, immunoblotting, RNA sequencing, senescence and mitochondrial function assays, confocal imaging, immunohistochemical, mass spectrometry, and co-immunoprecipitation. In vivo, we used MAVS-deficient models combined with CD8<sup>+</sup> T-cell depletion, programmed cell death protein-1 (PD-1) blockade, or reactive oxygen species (ROS) scavenging by N-acetylcysteine (NAC), with immune infiltration characterized by flow cytometry.</p><p><strong>Results: </strong>Clinical evidence links elevated MAVS expression in renal tumors to poor prognosis and diminished CD8<sup>+</sup> T-cell infiltration. Strikingly, tumor-intrinsic MAVS deficiency curbed malignant progression by triggering cellular senescence and fostering a permissive niche for CD8<sup>+</sup> T-cell activation and recruitment. Mechanistically, MAVS orchestrates mitochondrial integrity by co-localizing with and stabilizing chemokine-like factor-like MARVEL transmembrane domain-containing 6 (CMTM6), thereby shielding it from lysosomal degradation. Disruption of this axis provoked mitochondrial dysfunction and ROS accumulation, culminating in senescence and an SASP marked by chemokine C-C motif ligand 3 (CCL3). Thus, despite dampening canonical innate immune signaling, MAVS deletion unleashed potent antitumor immunity via CCL3-mediated CD8<sup>+</sup> T-cell recruitment, an effect abolished by CD8<sup>+</sup> T-cell depletion or ROS scavenging with NAC. Leveraging this paradigm, we demonstrated that tumor-specific MAVS deficiency acts synergistically with PD-1 blockade to achieve robust therapeutic efficacy.</p><p><strong>Conclusions: </strong>Our findings establish the tumor-intrinsic MAVS/CMTM6/CCL3 axis as a previously unrecognized critical regulator of senescence-driven antitumor immunity in renal carcinoma. Therapeutic targeting of this axis presents a promising strategy to curtail tumor progression and potentiate immunotherapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766799/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862828","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}
引用次数: 0
Overcoming impaired antigen presentation in tumor-draining lymph nodes facilitates immunotherapy. 克服肿瘤引流淋巴结中受损的抗原呈递有助于免疫治疗。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-30 DOI: 10.1136/jitc-2025-013364
Meghan J O'Melia, Lutz Menzel, Pin-Ji Lei, Hengbo Zhou, Xingjian Zhang, Neian Contreras-Alvarado, Johanna J Rajotte, Lingshan Liu, Mohammad R Nikmaneshi, James W Baish, Jessalyn M Ubellacker, Genevieve M Boland, Sonia Cohen, Timothy P Padera, Lance L Munn

Background: Immunotherapies have revolutionized cancer care in recent decades, but approved therapies often fail and currently only target specific steps in the generation of anti-cancer immune responses. Notably, the majority of approved immunotherapies do not target antigen processing and presentation, which are key steps in the development of immune responses and harbor potential as targets to improve immunotherapy. Here, we hypothesize that tumor-mediated alterations in cytokine concentrations alter antigen presentation, which can be normalized by locoregional cytokine delivery or targeted immunological adjuvant delivery.

Methods: We used mouse models of breast cancer, with analysis by flow cytometry, immunofluorescence, confocal imaging, and single-cell RNA sequencing to address the impacts of tumors on locoregional antigen presentation, along with mechanisms to remedy these impacts.

Results: Here, we demonstrate that breast tumors induce locoregional impairments in dendritic cell antigen presentation that limits anti-cancer antigen-specific T cell responses. Antigen processing was not impaired in dendritic cells within the tumor-draining lymph node. A reduction of the cytokine IL-1β in tumor-draining lymph nodes was responsible for impairments in antigen presentation by dendritic cells. As such, we tested the ability of dendritic cells in lymph nodes at various distances from the primary tumor to be activated utilizing an antigen-agnostic immunological adjuvant delivery strategy. We observed improved antitumor T cell responses when immunological adjuvant was delivered to cancer antigen-positive lymph nodes distant from the tumor, suggesting that these lymph nodes can be targeted to improve anti-cancer immune responses. When combined with immune checkpoint blockade, delivery of immunological adjuvant to distant lymph nodes led to long-term survival and protection from recurrence. Antigen presentation by dendritic cells and T cell responses could also be recovered by exogenous delivery of IL-1β via intratumoral injection, with improved survival when combined with immune checkpoint blockade.

Conclusions: This study demonstrates that tumor-induced impairments in antigen presentation in tumor-draining lymph nodes can be overcome by the appropriate introduction of immunological adjuvant to tumor-distant lymph nodes or by restoring IL-1β to the tumor-draining lymph node. These strategies can induce high-quality, durable immune responses and have clinical implications for expanding the efficacy of immunotherapies.

背景:近几十年来,免疫疗法已经彻底改变了癌症治疗,但批准的疗法往往失败,目前仅针对产生抗癌免疫反应的特定步骤。值得注意的是,大多数已批准的免疫疗法并不针对抗原加工和呈递,而抗原加工和呈递是免疫反应发展的关键步骤,具有改善免疫治疗的潜力。在这里,我们假设肿瘤介导的细胞因子浓度的改变改变了抗原呈递,这可以通过局部细胞因子递送或靶向免疫佐剂递送来正常化。方法:我们使用乳腺癌小鼠模型,通过流式细胞术、免疫荧光、共聚焦成像和单细胞RNA测序分析肿瘤对局部抗原呈递的影响,以及修复这些影响的机制。结果:在这里,我们证明了乳腺肿瘤诱导树突状细胞抗原呈递的局部区域损伤,限制了抗癌抗原特异性T细胞反应。肿瘤引流淋巴结内树突状细胞的抗原加工未受损。肿瘤引流淋巴结中细胞因子IL-1β的减少是树突状细胞抗原呈递受损的原因。因此,我们测试了淋巴结中距离原发肿瘤不同距离的树突状细胞被激活的能力,利用抗原不可知的免疫佐剂递送策略。我们观察到,当免疫佐剂被递送到远离肿瘤的癌抗原阳性淋巴结时,抗肿瘤T细胞反应得到改善,这表明这些淋巴结可以靶向改善抗癌免疫反应。当与免疫检查点阻断联合使用时,向远处淋巴结输送免疫佐剂可导致长期生存和防止复发。树突状细胞和T细胞反应的抗原呈递也可以通过肿瘤内注射外源性IL-1β来恢复,当结合免疫检查点阻断时,生存率提高。结论:本研究表明,肿瘤诱导的肿瘤引流淋巴结抗原呈递损伤可以通过向肿瘤远端淋巴结适当引入免疫佐剂或通过将IL-1β恢复到肿瘤引流淋巴结来克服。这些策略可以诱导高质量、持久的免疫反应,并对扩大免疫疗法的疗效具有临床意义。
{"title":"Overcoming impaired antigen presentation in tumor-draining lymph nodes facilitates immunotherapy.","authors":"Meghan J O'Melia, Lutz Menzel, Pin-Ji Lei, Hengbo Zhou, Xingjian Zhang, Neian Contreras-Alvarado, Johanna J Rajotte, Lingshan Liu, Mohammad R Nikmaneshi, James W Baish, Jessalyn M Ubellacker, Genevieve M Boland, Sonia Cohen, Timothy P Padera, Lance L Munn","doi":"10.1136/jitc-2025-013364","DOIUrl":"10.1136/jitc-2025-013364","url":null,"abstract":"<p><strong>Background: </strong>Immunotherapies have revolutionized cancer care in recent decades, but approved therapies often fail and currently only target specific steps in the generation of anti-cancer immune responses. Notably, the majority of approved immunotherapies do not target antigen processing and presentation, which are key steps in the development of immune responses and harbor potential as targets to improve immunotherapy. Here, we hypothesize that tumor-mediated alterations in cytokine concentrations alter antigen presentation, which can be normalized by locoregional cytokine delivery or targeted immunological adjuvant delivery.</p><p><strong>Methods: </strong>We used mouse models of breast cancer, with analysis by flow cytometry, immunofluorescence, confocal imaging, and single-cell RNA sequencing to address the impacts of tumors on locoregional antigen presentation, along with mechanisms to remedy these impacts.</p><p><strong>Results: </strong>Here, we demonstrate that breast tumors induce locoregional impairments in dendritic cell antigen presentation that limits anti-cancer antigen-specific T cell responses. Antigen processing was not impaired in dendritic cells within the tumor-draining lymph node. A reduction of the cytokine IL-1β in tumor-draining lymph nodes was responsible for impairments in antigen presentation by dendritic cells. As such, we tested the ability of dendritic cells in lymph nodes at various distances from the primary tumor to be activated utilizing an antigen-agnostic immunological adjuvant delivery strategy. We observed improved antitumor T cell responses when immunological adjuvant was delivered to cancer antigen-positive lymph nodes distant from the tumor, suggesting that these lymph nodes can be targeted to improve anti-cancer immune responses. When combined with immune checkpoint blockade, delivery of immunological adjuvant to distant lymph nodes led to long-term survival and protection from recurrence. Antigen presentation by dendritic cells and T cell responses could also be recovered by exogenous delivery of IL-1β via intratumoral injection, with improved survival when combined with immune checkpoint blockade.</p><p><strong>Conclusions: </strong>This study demonstrates that tumor-induced impairments in antigen presentation in tumor-draining lymph nodes can be overcome by the appropriate introduction of immunological adjuvant to tumor-distant lymph nodes or by restoring IL-1β to the tumor-draining lymph node. These strategies can induce high-quality, durable immune responses and have clinical implications for expanding the efficacy of immunotherapies.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766806/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862953","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}
引用次数: 0
Affinity-matured CD72-targeting nanobody CAR T cells enhance elimination of antigen-low B-cell malignancies. 亲和力成熟的靶向cd72纳米体CAR - T细胞增强了低抗原b细胞恶性肿瘤的消除。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-30 DOI: 10.1136/jitc-2025-012013
Adila Izgutdina, Tasfia Rashid, William C Temple, Sarah Aminov, Bonell Patiño-Escobar, Sujata Walunj, Huimin Geng, Hiroyuki Takamatsu, Daniel Gil-Alós, Amrik S Kang, Emilio Ramos, Szu-Ying Chen, Haley Johnson, Matthew A Nix, Akul Naik, Mingcheng Li, Constance M Yuan, Hao-Wei Wang, Srabani Sahu, Rebecca C Larson, Christopher Carpenter, Fernando Salangsang, Paul Phojanakong, Juan Antonio Camara Serrano, Isa Tariq, Ons Zakraoui, Veronica Steri, Antonio Valeri, Joaquin Martinez-Lopez, Marcela V Maus, Samir Parekh, Amit Verma, Nirali N Shah, Arun P Wiita

Background: Chimeric antigen receptor (CAR) T-cell therapies are highly efficacious for several different hematologic cancers. However, for most CAR T targets it is observed that low surface antigen density on tumors can significantly reduce therapeutic efficacy. In this study, we explore this dynamic in the context of CD72, a surface antigen we recently found as a promising target for refractory B-cell cancers, but for which CD72 low antigen density can lead to therapeutic resistance in preclinical models.

Methods: Primary samples were accessed via institutional review board-approved protocols. Affinity-matured and humanized nanobody clones were previously described in Temple et al. (2023). CAR T cells were generated via lentiviral transduction. In vitro cytotoxicity assays were performed using luciferase-labeled cell lines. In vivo studies were performed using cell line-derived or patient-derived xenografts implanted in NOD scid gamma mice.

Results: We first confirmed ubiquitous CD72 expression across a range of primary B-cell non-Hodgkin lymphomas. We further found that after resistance to CD19-directed therapies, across both B-cell acute lymphoblastic leukemia (B-ALL) models and primary tumor samples, surface CD72 expression was largely preserved while CD22 expression was significantly diminished. Affinity maturation of a nanobody targeting CD72, when incorporated into CAR T cells, led to more effective elimination in vitro of isogenic models of CD72 low-expressing tumors. These results suggested that nanobody-based CAR T cells (nanoCARs) may exhibit a similar relationship between binder affinity, antigen expression, and efficacy as previously demonstrated only for single chain variable fragment-based CAR T cells. Surprisingly, however, this significantly improved in vitro efficacy only translated to modest in vivo survival benefit. As a parallel strategy to enhance CAR T function, we found that the small molecule bryostatin could also significantly increase CD72 surface antigen density on B-cell malignancy models. Structural modeling and biochemical analysis identified critical residues improving CD72 antigen recognition of our lead affinity-matured nanobody.

Conclusions: Together, these findings support affinity-matured CD72 nanoCARs as a potential immunotherapy product for CD19-refractory B-cell cancers. Our results also suggest that for B-ALL in particular, CD72 may be a preferable second-line immunotherapy target over CD22.

背景:嵌合抗原受体(CAR) t细胞疗法对几种不同的血液病非常有效。然而,对于大多数CAR - T靶点,观察到肿瘤表面抗原密度低会显著降低治疗效果。在这项研究中,我们在CD72的背景下探索了这种动态,CD72是一种表面抗原,我们最近发现它是难治性b细胞癌的一个有希望的靶标,但在临床前模型中,CD72低抗原密度可能导致治疗耐药。方法:通过机构审查委员会批准的方案获取主要样本。亲和成熟和人源化的纳米体克隆先前在Temple等人(2023)中描述过。CAR - T细胞通过慢病毒转导产生。使用荧光素酶标记的细胞系进行体外细胞毒性测定。体内研究使用细胞系来源或患者来源的异种移植物植入NOD scid γ小鼠。结果:我们首次证实了CD72在一系列原发性b细胞非霍奇金淋巴瘤中的普遍表达。我们进一步发现,在对cd19靶向治疗产生耐药性后,在b细胞急性淋巴细胞白血病(B-ALL)模型和原发肿瘤样本中,表面CD72表达基本保留,而CD22表达显著降低。靶向CD72的纳米体的亲和成熟,当整合到CAR - T细胞中时,导致体外更有效地消除CD72低表达肿瘤的等基因模型。这些结果表明,基于纳米体的CAR - T细胞(nanoCARs)可能在结合物亲和力、抗原表达和功效之间表现出类似的关系,而之前仅在基于单链可变片段的CAR - T细胞中证实了这一点。然而,令人惊讶的是,这种显著提高的体外疗效仅转化为适度的体内生存益处。作为增强CAR - T功能的平行策略,我们发现小分子苔藓抑素也可以显著增加b细胞恶性肿瘤模型上的CD72表面抗原密度。结构建模和生化分析确定了提高我们的铅亲和成熟纳米体CD72抗原识别的关键残基。结论:总之,这些发现支持亲和成熟的CD72纳米car作为cd19难治性b细胞癌的潜在免疫治疗产品。我们的研究结果还表明,特别是对于B-ALL, CD72可能是比CD22更好的二线免疫治疗靶点。
{"title":"Affinity-matured CD72-targeting nanobody CAR T cells enhance elimination of antigen-low B-cell malignancies.","authors":"Adila Izgutdina, Tasfia Rashid, William C Temple, Sarah Aminov, Bonell Patiño-Escobar, Sujata Walunj, Huimin Geng, Hiroyuki Takamatsu, Daniel Gil-Alós, Amrik S Kang, Emilio Ramos, Szu-Ying Chen, Haley Johnson, Matthew A Nix, Akul Naik, Mingcheng Li, Constance M Yuan, Hao-Wei Wang, Srabani Sahu, Rebecca C Larson, Christopher Carpenter, Fernando Salangsang, Paul Phojanakong, Juan Antonio Camara Serrano, Isa Tariq, Ons Zakraoui, Veronica Steri, Antonio Valeri, Joaquin Martinez-Lopez, Marcela V Maus, Samir Parekh, Amit Verma, Nirali N Shah, Arun P Wiita","doi":"10.1136/jitc-2025-012013","DOIUrl":"10.1136/jitc-2025-012013","url":null,"abstract":"<p><strong>Background: </strong>Chimeric antigen receptor (CAR) T-cell therapies are highly efficacious for several different hematologic cancers. However, for most CAR T targets it is observed that low surface antigen density on tumors can significantly reduce therapeutic efficacy. In this study, we explore this dynamic in the context of CD72, a surface antigen we recently found as a promising target for refractory B-cell cancers, but for which CD72 low antigen density can lead to therapeutic resistance in preclinical models.</p><p><strong>Methods: </strong>Primary samples were accessed via institutional review board-approved protocols. Affinity-matured and humanized nanobody clones were previously described in Temple <i>et al.</i> (2023). CAR T cells were generated via lentiviral transduction. In vitro cytotoxicity assays were performed using luciferase-labeled cell lines. In vivo studies were performed using cell line-derived or patient-derived xenografts implanted in NOD <i>scid</i> gamma mice.</p><p><strong>Results: </strong>We first confirmed ubiquitous CD72 expression across a range of primary B-cell non-Hodgkin lymphomas. We further found that after resistance to CD19-directed therapies, across both B-cell acute lymphoblastic leukemia (B-ALL) models and primary tumor samples, surface CD72 expression was largely preserved while CD22 expression was significantly diminished. Affinity maturation of a nanobody targeting CD72, when incorporated into CAR T cells, led to more effective elimination in vitro of isogenic models of CD72 low-expressing tumors. These results suggested that nanobody-based CAR T cells (nanoCARs) may exhibit a similar relationship between binder affinity, antigen expression, and efficacy as previously demonstrated only for single chain variable fragment-based CAR T cells. Surprisingly, however, this significantly improved in vitro efficacy only translated to modest in vivo survival benefit. As a parallel strategy to enhance CAR T function, we found that the small molecule bryostatin could also significantly increase CD72 surface antigen density on B-cell malignancy models. Structural modeling and biochemical analysis identified critical residues improving CD72 antigen recognition of our lead affinity-matured nanobody.</p><p><strong>Conclusions: </strong>Together, these findings support affinity-matured CD72 nanoCARs as a potential immunotherapy product for CD19-refractory B-cell cancers. Our results also suggest that for B-ALL in particular, CD72 may be a preferable second-line immunotherapy target over CD22.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12766774/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862700","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}
引用次数: 0
Genetic engineering and allogeneic optimization of Vδ1 γδ CAR-T cells (ADI-270) for cancer immunotherapy. 用于肿瘤免疫治疗的Vδ1 γδ CAR-T细胞(ADI-270)的基因工程和异体优化
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-25 DOI: 10.1136/jitc-2025-013537
Wenlong Liu, Branden S Moriarity, Beau R Webber

Genetic engineering has fundamentally transformed T cell-based therapies by enabling tumor targeting capability, improving their functionality, and facilitating allogeneic use. These strategies-originally developed in αβ chimeric antigen receptor (CAR)-T cells-have become increasingly established as blueprints for enhancing the function of other immune effector cells, including gamma delta (γδ) T cells. A recent study by Nishimoto et al showcased the adaptation of these engineering approaches to Vδ1 γδ T cells (ADI-270) by coexpressing a CD70-targeted CAR and a dominant-negative TGFβRII receptor (dnTGFβRII) to target CD70+ malignancies, addressing immunosuppression and host-versus-graft rejection. This commentary explores αβ T cell-derived engineering strategies applicable to γδ T cells, while also highlighting genome-editing innovations poised to advance next-generation γδ CAR-T development.

基因工程已经从根本上改变了基于T细胞的治疗方法,使肿瘤靶向能力,提高其功能,并促进同种异体的使用。这些策略最初是在αβ嵌合抗原受体(CAR)-T细胞中开发的,现已越来越多地作为增强其他免疫效应细胞(包括γδ (γδ) T细胞)功能的蓝图。Nishimoto等人最近的一项研究表明,通过共表达CD70靶向CAR和显性阴性TGFβRII受体(dnTGFβRII),这些工程方法适用于Vδ1 γδ T细胞(ADI-270),以靶向CD70+恶性肿瘤,解决免疫抑制和宿主抗移植排斥反应。这篇评论探讨了适用于γδ T细胞的αβ T细胞衍生的工程策略,同时也强调了基因组编辑创新,有望推进下一代γδ CAR-T的发展。
{"title":"Genetic engineering and allogeneic optimization of Vδ1 γδ CAR-T cells (ADI-270) for cancer immunotherapy.","authors":"Wenlong Liu, Branden S Moriarity, Beau R Webber","doi":"10.1136/jitc-2025-013537","DOIUrl":"10.1136/jitc-2025-013537","url":null,"abstract":"<p><p>Genetic engineering has fundamentally transformed T cell-based therapies by enabling tumor targeting capability, improving their functionality, and facilitating allogeneic use. These strategies-originally developed in αβ chimeric antigen receptor (CAR)-T cells-have become increasingly established as blueprints for enhancing the function of other immune effector cells, including gamma delta (γδ) T cells. A recent study by Nishimoto <i>et al</i> showcased the adaptation of these engineering approaches to Vδ1 γδ T cells (ADI-270) by coexpressing a CD70-targeted CAR and a dominant-negative TGFβRII receptor (dnTGFβRII) to target CD70<sup>+</sup> malignancies, addressing immunosuppression and host-versus-graft rejection. This commentary explores αβ T cell-derived engineering strategies applicable to γδ T cells, while also highlighting genome-editing innovations poised to advance next-generation γδ CAR-T development.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 12","pages":""},"PeriodicalIF":10.6,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12742126/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145843735","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}
引用次数: 0
期刊
Journal for Immunotherapy of Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1