首页 > 最新文献

Cancer immunology research最新文献

英文 中文
Seeing First: Introducing the Resource Report at Cancer Immunology Research. 先看:介绍癌症免疫学研究的资源报告。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-1175
Evan W Newell
{"title":"Seeing First: Introducing the Resource Report at Cancer Immunology Research.","authors":"Evan W Newell","doi":"10.1158/2326-6066.CIR-25-1175","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-1175","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"13 11","pages":"1696-1697"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145430355","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
Chronic ISG15 Exposure Accelerates CD8+ T-cell Dysfunction while Increasing PD-1 Blockade Sensitivity in Oral Squamous Cell Carcinoma. 慢性ISG15暴露在口腔鳞状细胞癌中加速CD8+ t细胞功能障碍,同时增加PD-1阻断敏感性。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-0047
Yu-Lin Chen, Amir Yousif, Chung-Hsing Chen, Ava Lowin, Abbey A Saadey, Ssu-Han Wang, Shih Sheng Jiang, Ya-Wen Chen, Hazem E Ghoneim

Immunotherapy has emerged as a promising treatment for head and neck squamous cell carcinoma, yet clinical responses remain limited. Elevated expression of interferon-stimulated gene 15 (ISG15), commonly observed in oral squamous cell carcinoma (OSCC), may contribute to this limited efficacy. Although chronic interferon signaling is known to impair CD8+ T-cell function, the specific role of secreted ISG15 in T-cell exhaustion remains unclear. In this study, we report that the analysis of human OSCC datasets revealed significant enrichment of the ISG core score, including ISG15, in tumors compared with adjacent nontumor tissues. Using an in vitro model of human T-cell dysfunction, we found that acute ISG15 exposure enhanced CD8+ T-cell effector functions, whereas prolonged exposure induced severe dysfunction via a CD11a/LFA-1-independent, endocytosis-dependent mechanism. In an immunocompetent orthotopic OSCC model, ISG15-expressing tumors exhibited accelerated growth and recruited more tumor-reactive CD8+ T cells; however, these cells were functionally impaired. Moreover, PD-1 blockade treatment significantly slowed tumor progression and restored T-cell function in ISG15-expressing tumors. Together, our findings reveal that chronic ISG15 exposure promotes CD8+ T-cell dysfunction, but these cells remain responsive to PD-1 blockade. Furthermore, this study identifies ISG15 as a potential biomarker for identifying patients who are likely to benefit from PD-1 blockade therapy.

免疫疗法已成为头颈部鳞状细胞癌(HNSCC)的一种有希望的治疗方法,但临床反应仍然有限。在口腔鳞状细胞癌(OSCC)中常见的干扰素刺激基因15 (ISG15)表达升高可能是导致这种有限疗效的原因。虽然已知慢性干扰素信号会损害CD8+ T细胞功能,但分泌的ISG15在T细胞衰竭中的具体作用尚不清楚。对人类OSCC数据集的分析显示,与邻近的非肿瘤组织相比,肿瘤中ISG核心评分(包括isg15)显著富集。利用一种新的体外人类T细胞功能障碍模型,我们发现急性暴露于ISG15可增强CD8+ T细胞效应功能,而长期暴露于ISG15可通过CD11a/ lfa -1不依赖、内吞依赖的机制诱导严重功能障碍。在免疫耐受的原位OSCC模型中,表达isg15的肿瘤表现出加速生长和募集更多肿瘤反应性CD8+ T细胞;然而,这些细胞功能受损。重要的是,PD-1阻断治疗显著减缓了表达isg15的肿瘤的肿瘤进展并恢复了T细胞功能。总之,我们的研究结果表明,慢性ISG15暴露促进CD8+ T细胞功能障碍,但这些细胞仍然对PD-1阻断有反应。本研究确定ISG15是一种潜在的生物标志物,可用于识别可能受益于PD-1阻断治疗的患者。
{"title":"Chronic ISG15 Exposure Accelerates CD8+ T-cell Dysfunction while Increasing PD-1 Blockade Sensitivity in Oral Squamous Cell Carcinoma.","authors":"Yu-Lin Chen, Amir Yousif, Chung-Hsing Chen, Ava Lowin, Abbey A Saadey, Ssu-Han Wang, Shih Sheng Jiang, Ya-Wen Chen, Hazem E Ghoneim","doi":"10.1158/2326-6066.CIR-25-0047","DOIUrl":"10.1158/2326-6066.CIR-25-0047","url":null,"abstract":"<p><p>Immunotherapy has emerged as a promising treatment for head and neck squamous cell carcinoma, yet clinical responses remain limited. Elevated expression of interferon-stimulated gene 15 (ISG15), commonly observed in oral squamous cell carcinoma (OSCC), may contribute to this limited efficacy. Although chronic interferon signaling is known to impair CD8+ T-cell function, the specific role of secreted ISG15 in T-cell exhaustion remains unclear. In this study, we report that the analysis of human OSCC datasets revealed significant enrichment of the ISG core score, including ISG15, in tumors compared with adjacent nontumor tissues. Using an in vitro model of human T-cell dysfunction, we found that acute ISG15 exposure enhanced CD8+ T-cell effector functions, whereas prolonged exposure induced severe dysfunction via a CD11a/LFA-1-independent, endocytosis-dependent mechanism. In an immunocompetent orthotopic OSCC model, ISG15-expressing tumors exhibited accelerated growth and recruited more tumor-reactive CD8+ T cells; however, these cells were functionally impaired. Moreover, PD-1 blockade treatment significantly slowed tumor progression and restored T-cell function in ISG15-expressing tumors. Together, our findings reveal that chronic ISG15 exposure promotes CD8+ T-cell dysfunction, but these cells remain responsive to PD-1 blockade. Furthermore, this study identifies ISG15 as a potential biomarker for identifying patients who are likely to benefit from PD-1 blockade therapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1829-1844"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144943937","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
Tissue-Specific Immunosuppressive and Proliferating Macrophages Fuel Early Metastatic Progression of Human Colorectal Cancer to the Liver. 组织特异性免疫抑制和增殖巨噬细胞促进人类结直肠癌向肝脏的早期转移进展。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-0031
Paolo Marzano, Cristiana Soldani, Valentina Cazzetta, Barbara Franceschini, Sara Terzoli, Anna Carletti, Michela Anna Polidoro, Federica Marchesi, Massimo Locati, Gianluca Basso, Ana Lleo, Guido Costa, Guido Torzilli, Flavio Milana, Rocco Piazza, Paola Spaggiari, Luca Di Tommaso, Joanna Mikulak, Domenico Mavilio, Matteo Donadon

Early synchronous colorectal liver metastasis (CRLM) represents a clinical condition characterized by the simultaneous presence of primary colorectal cancer and metastatic liver lesions. In this study, we characterized the tissue-specific transcriptomes, phenotypes, and functional relevance of tumor-associated macrophages (TAM) within the tumor microenvironment (TME) of colorectal cancer and CRLM specimens from patients who underwent simultaneous surgical removal of these malignancies. The high-throughput single-cell transcriptional analysis revealed an inverse ratio of inflammatory and immunoregulatory TAMs in the colorectal cancer and CRLM TMEs, along with heterogeneity in both tumoral tissues. Furthermore, we found that inflammatory TAMs in colorectal cancer expressed inhibitory ligands that might support immune escape, thus favoring liver metastatic progression. In contrast, CRLM lesions possessed a highly immunosuppressive milieu characterized by large proliferative CTLA4+ immunoregulatory TAMs and the presence of IL7R+ cytotoxic TAMs. Higher frequencies of these specific TAM subsets in CRLM were associated with shorter disease-free survival and worse patient prognosis. The identification and characterization of immunoregulatory TAMs preferentially enriched in CRLM is key for the development of novel immunotherapeutic strategies aimed at boosting anticancer immune responses within the TME.

早期同步性结肝转移(CRLM)是一种以原发性结直肠癌(CRC)和转移性肝脏病变同时存在为特征的临床状态。在这项研究中,我们对同时接受手术切除这些恶性肿瘤的CRC和CRLM标本的肿瘤微环境(TME)中肿瘤相关巨噬细胞(tam)的组织特异性转录组、表型和功能相关性进行了表征。高通量单细胞转录分析显示,CRC和CRLM TMEs中炎症性和免疫调节性tam呈反比,并且在两种肿瘤组织中均存在异质性。此外,我们发现CRC中的炎性tam表达抑制配体,可能支持免疫逃逸,从而促进肝转移进展。相比之下,CRLM病变具有高度免疫抑制的环境,其特征是大量增殖性CTLA4+免疫调节性tam和IL7R+细胞毒性tam的存在。CRLM中这些特异性TAM亚群的较高频率与较短的无病生存期和较差的患者预后相关。鉴定和鉴定优先富集于CRLM的免疫调节tam是开发旨在增强TME内抗癌免疫应答的新型免疫治疗策略的关键。
{"title":"Tissue-Specific Immunosuppressive and Proliferating Macrophages Fuel Early Metastatic Progression of Human Colorectal Cancer to the Liver.","authors":"Paolo Marzano, Cristiana Soldani, Valentina Cazzetta, Barbara Franceschini, Sara Terzoli, Anna Carletti, Michela Anna Polidoro, Federica Marchesi, Massimo Locati, Gianluca Basso, Ana Lleo, Guido Costa, Guido Torzilli, Flavio Milana, Rocco Piazza, Paola Spaggiari, Luca Di Tommaso, Joanna Mikulak, Domenico Mavilio, Matteo Donadon","doi":"10.1158/2326-6066.CIR-25-0031","DOIUrl":"10.1158/2326-6066.CIR-25-0031","url":null,"abstract":"<p><p>Early synchronous colorectal liver metastasis (CRLM) represents a clinical condition characterized by the simultaneous presence of primary colorectal cancer and metastatic liver lesions. In this study, we characterized the tissue-specific transcriptomes, phenotypes, and functional relevance of tumor-associated macrophages (TAM) within the tumor microenvironment (TME) of colorectal cancer and CRLM specimens from patients who underwent simultaneous surgical removal of these malignancies. The high-throughput single-cell transcriptional analysis revealed an inverse ratio of inflammatory and immunoregulatory TAMs in the colorectal cancer and CRLM TMEs, along with heterogeneity in both tumoral tissues. Furthermore, we found that inflammatory TAMs in colorectal cancer expressed inhibitory ligands that might support immune escape, thus favoring liver metastatic progression. In contrast, CRLM lesions possessed a highly immunosuppressive milieu characterized by large proliferative CTLA4+ immunoregulatory TAMs and the presence of IL7R+ cytotoxic TAMs. Higher frequencies of these specific TAM subsets in CRLM were associated with shorter disease-free survival and worse patient prognosis. The identification and characterization of immunoregulatory TAMs preferentially enriched in CRLM is key for the development of novel immunotherapeutic strategies aimed at boosting anticancer immune responses within the TME.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1783-1797"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580778/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144871625","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
Tumor-Derived EBI3 Promotes CD8+ T-cell Exhaustion via STAT4-IL10/CCL5 in Gastric Cancer. 肿瘤来源的EBI3在胃癌中通过STAT4-IL-10/CCL5促进CD8+ T细胞衰竭。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-24-1228
Yong-Jia Yan, Xin Liu, Daohan Wang, Zexuan Shen, Wenxin Zhang, Zhaoxiong Zhang, Yangpu Jia, Peiyao Wang, Yuman Fong, Yanghee Woo, Weihua Fu

Combination chemotherapy and immunotherapy are effective against advanced gastric cancer. However, T-cell exhaustion in the tumor microenvironment may decrease the immune response and compromise the effectiveness of immunotherapy. Herein, we report the potential role of EBI3 in promoting T-cell exhaustion and its mechanism in gastric cancer, showing high expression of EBI3 in gastric cancer. Correlation analysis between EBI3 expression level and clinical-pathologic features indicated significant associations with tumor stage, nodal staging, pathologic stage, and degree of tumor differentiation. EBI3 expression levels correlated with a state of high CD8+ T-cell exhaustion, as identified by transcriptome sequencing and mouse orthotopic gastric cancer models. On exposure to EBI3, CD8+ T cells showed signs of cell exhaustion as reduced cytokine secretion and increased expression of inhibitory receptors in in vitro/vivo studies. Mechanistically, EBI3 induced T-cell exhaustion by promoting the phosphorylation of STAT4 and upregulating the transcription of downstream target genes CCL5 and IL10. An anti-EBI3 heptapeptide (Val-Tyr-Leu-His-Trp-His-Asp) was developed, which competitively bound EBI3 and reversed the induction of T-cell exhaustion. Taken together, we identified a T-cell exhaustion mechanism in gastric cancer via the EBI3-STAT4-IL10/CCL5 axis and developed an anti-EBI3 heptapeptide with an antagonistic function. These findings provide a potential immunotherapeutic target and support the development of EBI3-based interventions to enhance immunotherapy efficacy in gastric cancer.

化疗联合免疫治疗是治疗晚期胃癌的有效方法。然而,肿瘤微环境中的T细胞耗竭可能降低免疫应答,损害免疫治疗的有效性。本文报道了EBI3在胃癌中促进T细胞衰竭的潜在作用及其机制,显示了EBI3在胃癌中的高表达。EBI3表达水平与临床病理特征的相关性分析显示,EBI3表达水平与肿瘤分期、淋巴结分期、病理分期、肿瘤分化程度有显著相关性。通过转录组测序和小鼠原位GC模型发现,EBI3表达水平与CD8+ T细胞高耗竭状态相关。在体外/体内研究中,暴露于EBI3后,CD8+ T细胞表现出细胞衰竭的迹象,细胞因子分泌减少,抑制受体表达增加。在机制上,EBI3通过促进STAT4的磷酸化,上调下游靶基因CCL5和IL-10的转录来诱导T细胞衰竭。我们开发了一种抗EBI3七肽(val - tir - leu - his - trp - his - asp),它竞争性地结合EBI3并逆转诱导的T细胞衰竭。综上所述,我们通过EBI3-STAT4-IL10/CCL5轴确定了GC中的T细胞衰竭机制,并开发了具有拮抗功能的抗ebi3七肽。这些发现提供了一个潜在的免疫治疗靶点,并支持基于ebi3的干预措施的发展,以提高GC的免疫治疗效果。
{"title":"Tumor-Derived EBI3 Promotes CD8+ T-cell Exhaustion via STAT4-IL10/CCL5 in Gastric Cancer.","authors":"Yong-Jia Yan, Xin Liu, Daohan Wang, Zexuan Shen, Wenxin Zhang, Zhaoxiong Zhang, Yangpu Jia, Peiyao Wang, Yuman Fong, Yanghee Woo, Weihua Fu","doi":"10.1158/2326-6066.CIR-24-1228","DOIUrl":"10.1158/2326-6066.CIR-24-1228","url":null,"abstract":"<p><p>Combination chemotherapy and immunotherapy are effective against advanced gastric cancer. However, T-cell exhaustion in the tumor microenvironment may decrease the immune response and compromise the effectiveness of immunotherapy. Herein, we report the potential role of EBI3 in promoting T-cell exhaustion and its mechanism in gastric cancer, showing high expression of EBI3 in gastric cancer. Correlation analysis between EBI3 expression level and clinical-pathologic features indicated significant associations with tumor stage, nodal staging, pathologic stage, and degree of tumor differentiation. EBI3 expression levels correlated with a state of high CD8+ T-cell exhaustion, as identified by transcriptome sequencing and mouse orthotopic gastric cancer models. On exposure to EBI3, CD8+ T cells showed signs of cell exhaustion as reduced cytokine secretion and increased expression of inhibitory receptors in in vitro/vivo studies. Mechanistically, EBI3 induced T-cell exhaustion by promoting the phosphorylation of STAT4 and upregulating the transcription of downstream target genes CCL5 and IL10. An anti-EBI3 heptapeptide (Val-Tyr-Leu-His-Trp-His-Asp) was developed, which competitively bound EBI3 and reversed the induction of T-cell exhaustion. Taken together, we identified a T-cell exhaustion mechanism in gastric cancer via the EBI3-STAT4-IL10/CCL5 axis and developed an anti-EBI3 heptapeptide with an antagonistic function. These findings provide a potential immunotherapeutic target and support the development of EBI3-based interventions to enhance immunotherapy efficacy in gastric cancer.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1873-1886"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144943996","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
Targeting Myeloid Cells for Cancer Immunotherapy. 靶向骨髓细胞用于癌症免疫治疗。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-0159
Lucas Blanchard, Andrew Mijacika, Juan C Osorio

Myeloid cells-including monocytes, macrophages, dendritic cells, and granulocytes-are critical architects of the tumor microenvironment, in which they exert diverse functions ranging from immunosuppressive to immunostimulatory. Advances in single-cell omics and high-dimensional immune profiling have unveiled the remarkable heterogeneity and plasticity of these cells, revealing lineage-specialized functions that shape cancer immunity. These discoveries have sparked growing interest in therapeutically targeting myeloid cells as a next-generation strategy in cancer immunotherapy. As a complementary or alternative approach to T cell-centered immunotherapies, myeloid-directed therapies offer unique opportunities to reprogram the immune landscape, enhance antitumor responses, and overcome resistance mechanisms. In this review, we highlight recent discoveries in myeloid cell biology in cancer and discuss emerging therapeutic targets, with an emphasis on antibody-based therapies that have reached clinical development. We further provide perspective on translational challenges to implement these approaches into the clinic and discuss how Fc-engineering and rational antibody design can optimize myeloid cell engagement and amplify their immune effector functions. Together, these advances position myeloid-directed immunotherapies as a promising approach to enhance the efficacy and durability of cancer treatment.

骨髓细胞——包括单核细胞、巨噬细胞、树突状细胞和粒细胞——是肿瘤微环境的关键构建者,它们发挥着从免疫抑制到免疫刺激的多种功能。单细胞组学和高维免疫图谱的进展揭示了这些细胞的显著异质性和可塑性,揭示了形成癌症免疫的谱系特异性功能。这些发现激发了人们对治疗靶向骨髓细胞作为下一代癌症免疫治疗策略的兴趣。作为T细胞中心免疫治疗的补充或替代方法,髓系定向治疗提供了重新编程免疫景观,增强抗肿瘤反应和克服耐药机制的独特机会。在这篇综述中,我们重点介绍了骨髓细胞生物学在癌症中的最新发现,并讨论了新兴的治疗靶点,重点是已经达到临床发展的基于抗体的治疗方法。我们进一步提供了将这些方法应用于临床的翻译挑战的观点,并讨论了fc工程和合理的抗体设计如何优化髓细胞接合并增强其免疫效应功能。总之,这些进展使骨髓定向免疫疗法成为提高癌症治疗疗效和持久性的一种有希望的方法。
{"title":"Targeting Myeloid Cells for Cancer Immunotherapy.","authors":"Lucas Blanchard, Andrew Mijacika, Juan C Osorio","doi":"10.1158/2326-6066.CIR-25-0159","DOIUrl":"10.1158/2326-6066.CIR-25-0159","url":null,"abstract":"<p><p>Myeloid cells-including monocytes, macrophages, dendritic cells, and granulocytes-are critical architects of the tumor microenvironment, in which they exert diverse functions ranging from immunosuppressive to immunostimulatory. Advances in single-cell omics and high-dimensional immune profiling have unveiled the remarkable heterogeneity and plasticity of these cells, revealing lineage-specialized functions that shape cancer immunity. These discoveries have sparked growing interest in therapeutically targeting myeloid cells as a next-generation strategy in cancer immunotherapy. As a complementary or alternative approach to T cell-centered immunotherapies, myeloid-directed therapies offer unique opportunities to reprogram the immune landscape, enhance antitumor responses, and overcome resistance mechanisms. In this review, we highlight recent discoveries in myeloid cell biology in cancer and discuss emerging therapeutic targets, with an emphasis on antibody-based therapies that have reached clinical development. We further provide perspective on translational challenges to implement these approaches into the clinic and discuss how Fc-engineering and rational antibody design can optimize myeloid cell engagement and amplify their immune effector functions. Together, these advances position myeloid-directed immunotherapies as a promising approach to enhance the efficacy and durability of cancer treatment.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1700-1715"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12477725/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944006","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
Evaluation of Proton Minibeam Radiotherapy on Antitumor Immune Responses in a Rat Model of Glioblastoma. 质子微束放射治疗对胶质母细胞瘤模型大鼠抗肿瘤免疫反应的评价。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-24-0902
Lorea Iturri, Miriam Riquelme-Perez, Pierre-Emannuel Bonté, Sarah Potiron, Christel Goudot, Marjorie Juchaux, Elise Brisebard, Cristèle Gilbert, Julie Espenon, Ramón Ortiz, Annalisa Patriarca, Ludovic De Marzi, Sebastián Amigorena, Yolanda Prezado

Treating radioresistant tumors like glioblastoma multiforme remains a challenge exacerbated by their immunosuppressive nature. Radiotherapy (RT) plays an immunomodulatory role, exerting both immunosuppressive and immunostimulatory effects. The nature of these effects depends on the total dose, dose per fraction, dose delivery method, and treatment length. Hypofractionation is observed to tip the balance toward immune stimulation. However, the use of hypofractionation is restricted in bulky tumors, such as gliomas, because of the high risk of toxicity. Therefore, finding new strategies leading to more favorable immune responses while reducing normal tissue toxicities could improve cancer treatment. In this study, we examine antitumoral immune responses to proton minibeam RT (pMBRT). Its immunomodulatory effects are not fully understood. To explore this, we conducted an in-depth characterization of the immune response to a curative dose of pMBRT in a preclinical orthotopic rat model of glioblastoma. Our findings revealed a close association between pMBRT and the immune response. pMBRT increased lymphocyte density in tumors more effectively than conventional proton therapy. Single-cell transcriptomics identified several immune cell types and unique transcriptional changes in tumor immune cells following pMBRT, including increased antibody production, chemotactic cytokine expression, and IFN responses. These results underscore the critical role of adaptive immunity, specifically T cells, in pMBRT's mechanism. The potential of pMBRT to trigger an antitumor immune response in a single RT session with minimal damage to healthy tissue makes it a promising candidate for future clinical trials and radioimmunotherapy combinations.

治疗放射耐药肿瘤如多形性胶质母细胞瘤(GBM)仍然是一个挑战,加剧了其免疫抑制的性质。放射治疗具有免疫调节作用,既具有免疫抑制作用,又具有免疫刺激作用。效果的性质取决于总剂量、每部分剂量、给药方法和治疗时间。观察到低分割使平衡倾向于免疫刺激。然而,由于毒性风险高,在体积较大的肿瘤(如胶质瘤)中使用低分割术受到限制。因此,寻找新的策略,导致更有利的免疫反应,同时减少正常组织的毒性,可以改善癌症治疗。在这里,我们研究质子微束放射治疗(pMBRT)的抗肿瘤免疫反应。然而,其免疫调节作用尚不完全清楚。为了探讨这一点,我们在临床前原位胶质母细胞瘤大鼠模型中对治疗剂量pMBRT的免疫反应进行了深入的表征。我们的研究结果揭示了pMBRT与免疫反应之间的密切联系。pMBRT比常规质子治疗更有效地增加肿瘤淋巴细胞密度。单细胞转录组学鉴定了pmbrt后肿瘤免疫细胞的几种免疫细胞类型和独特的转录变化,包括抗体产生、趋化细胞因子表达和干扰素反应的增加。这些结果强调了适应性免疫,特别是T细胞在pMBRT机制中的关键作用。pMBRT在单次放射治疗中触发抗肿瘤免疫反应的潜力,对健康组织的损伤最小,使其成为未来临床试验和放射免疫治疗组合的有希望的候选者。
{"title":"Evaluation of Proton Minibeam Radiotherapy on Antitumor Immune Responses in a Rat Model of Glioblastoma.","authors":"Lorea Iturri, Miriam Riquelme-Perez, Pierre-Emannuel Bonté, Sarah Potiron, Christel Goudot, Marjorie Juchaux, Elise Brisebard, Cristèle Gilbert, Julie Espenon, Ramón Ortiz, Annalisa Patriarca, Ludovic De Marzi, Sebastián Amigorena, Yolanda Prezado","doi":"10.1158/2326-6066.CIR-24-0902","DOIUrl":"10.1158/2326-6066.CIR-24-0902","url":null,"abstract":"<p><p>Treating radioresistant tumors like glioblastoma multiforme remains a challenge exacerbated by their immunosuppressive nature. Radiotherapy (RT) plays an immunomodulatory role, exerting both immunosuppressive and immunostimulatory effects. The nature of these effects depends on the total dose, dose per fraction, dose delivery method, and treatment length. Hypofractionation is observed to tip the balance toward immune stimulation. However, the use of hypofractionation is restricted in bulky tumors, such as gliomas, because of the high risk of toxicity. Therefore, finding new strategies leading to more favorable immune responses while reducing normal tissue toxicities could improve cancer treatment. In this study, we examine antitumoral immune responses to proton minibeam RT (pMBRT). Its immunomodulatory effects are not fully understood. To explore this, we conducted an in-depth characterization of the immune response to a curative dose of pMBRT in a preclinical orthotopic rat model of glioblastoma. Our findings revealed a close association between pMBRT and the immune response. pMBRT increased lymphocyte density in tumors more effectively than conventional proton therapy. Single-cell transcriptomics identified several immune cell types and unique transcriptional changes in tumor immune cells following pMBRT, including increased antibody production, chemotactic cytokine expression, and IFN responses. These results underscore the critical role of adaptive immunity, specifically T cells, in pMBRT's mechanism. The potential of pMBRT to trigger an antitumor immune response in a single RT session with minimal damage to healthy tissue makes it a promising candidate for future clinical trials and radioimmunotherapy combinations.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1854-1872"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580782/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144943886","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
Systematic Engineering of TROP2-Targeted CAR T-Cell Therapy Overcomes Resistance Pathways in Solid Tumors. trop2靶向CAR -t细胞治疗的系统工程克服了实体瘤的耐药途径。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-0527
Elliott J Brea, Simon Baldacci, Neil Savage, Francesco Facchinetti, Conor Hinchey, Sachiv Chakravarti, Alexis Mottram, Kenneth Ngo, Ha Vo, Brittaney A Leeper, Bishma Tuladhar, Suthakar Ganapathy, Elena V Ivanova, Aisha Saldanha, Marie-Anais Locquet, Abdulmajeed Salamah, Malcolm Holterhus, Evelyn B Mesler, Martina De Vizio, Carla Stornante, Marco Campisi, Navin R Mahadevan, Tran C Thai, Timothy J Haggerty, Zehua Li, Cui Nie, Changjing Deng, Xiaoxiao Wang, Louis L Liu, Thanh U Barbie, Prafulla C Gokhale, Cloud P Paweletz, Anusuya Ramasubramanian, Pasi A Jänne, David A Barbie, Eric L Smith

Antibody-based therapies have revolutionized cancer treatment but have several limitations. These include downregulation of the target antigen, mutation of the target epitope, and, in the case of antibody-drug conjugates (ADC), resistance to the chemotherapy warhead. As TROP2-targeted therapy with ADCs yields responses in TROP2+ solid tumors, but the responses lack the durability observed with other immunotherapy-based approaches, we developed TROP2-targeting chimeric antigen receptor (CAR) T cells as an alternative. The TROP2-directed CAR T cells showed high potency against multiple solid tumor models. Moreover, TROP2-directed CAR T-cell therapy preserved high potency in models of ADC resistance and could be further engineered to prevent cell therapy resistance. This was achieved by leveraging fully human single-domain (VH-only) binder discovery to rationally engineer dual epitope binding-based (biparatopic) CARs. This work highlights the potency of CAR T-cell therapies and how rational engineering leveraging dual-VH targeting domains can overcome resistance pathways to current therapies. In future work, the CAR engineering approaches presented here can serve as a platform to be partnered with other strategies to address the suppressive tumor microenvironment.

基于抗体的疗法已经彻底改变了癌症治疗,但也有一些局限性。这包括:靶抗原的下调;靶表位突变;或者在抗体药物偶联物(adc)的情况下,对化疗弹头的耐药性。由于adc靶向治疗TROP2在TROP2+实体瘤中产生应答,但缺乏其他基于免疫治疗方法观察到的持久性,我们开发了新的靶向TROP2的嵌合抗原受体(CAR) T细胞作为替代方案。我们观察到TROP2靶向CAR - T对多种实体瘤模型具有高效力。我们证明CAR - T细胞疗法可以在ADC耐药模型中保持高效力,并且可以进一步设计以防止细胞治疗耐药;利用全人类单域(VH-only)结合剂发现,合理地设计基于双表位结合(双异位)的car。这项工作强调了CAR - T细胞疗法的效力,以及利用双vh靶向结构域的合理工程如何克服对当前疗法的抗性途径。在未来的工作中,本文提出的CAR工程方法可以作为与其他策略合作的平台,以解决抑制性肿瘤微环境。这项工作强调了CAR - T细胞疗法的效力,以及利用双vh靶向结构域的合理工程如何克服对当前疗法的抗性途径。
{"title":"Systematic Engineering of TROP2-Targeted CAR T-Cell Therapy Overcomes Resistance Pathways in Solid Tumors.","authors":"Elliott J Brea, Simon Baldacci, Neil Savage, Francesco Facchinetti, Conor Hinchey, Sachiv Chakravarti, Alexis Mottram, Kenneth Ngo, Ha Vo, Brittaney A Leeper, Bishma Tuladhar, Suthakar Ganapathy, Elena V Ivanova, Aisha Saldanha, Marie-Anais Locquet, Abdulmajeed Salamah, Malcolm Holterhus, Evelyn B Mesler, Martina De Vizio, Carla Stornante, Marco Campisi, Navin R Mahadevan, Tran C Thai, Timothy J Haggerty, Zehua Li, Cui Nie, Changjing Deng, Xiaoxiao Wang, Louis L Liu, Thanh U Barbie, Prafulla C Gokhale, Cloud P Paweletz, Anusuya Ramasubramanian, Pasi A Jänne, David A Barbie, Eric L Smith","doi":"10.1158/2326-6066.CIR-25-0527","DOIUrl":"10.1158/2326-6066.CIR-25-0527","url":null,"abstract":"<p><p>Antibody-based therapies have revolutionized cancer treatment but have several limitations. These include downregulation of the target antigen, mutation of the target epitope, and, in the case of antibody-drug conjugates (ADC), resistance to the chemotherapy warhead. As TROP2-targeted therapy with ADCs yields responses in TROP2+ solid tumors, but the responses lack the durability observed with other immunotherapy-based approaches, we developed TROP2-targeting chimeric antigen receptor (CAR) T cells as an alternative. The TROP2-directed CAR T cells showed high potency against multiple solid tumor models. Moreover, TROP2-directed CAR T-cell therapy preserved high potency in models of ADC resistance and could be further engineered to prevent cell therapy resistance. This was achieved by leveraging fully human single-domain (VH-only) binder discovery to rationally engineer dual epitope binding-based (biparatopic) CARs. This work highlights the potency of CAR T-cell therapies and how rational engineering leveraging dual-VH targeting domains can overcome resistance pathways to current therapies. In future work, the CAR engineering approaches presented here can serve as a platform to be partnered with other strategies to address the suppressive tumor microenvironment.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1749-1763"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580781/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145013757","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
Targeting LxCxE Cleft Pocket of Retinoblastoma Protein in Immunosuppressive Macrophages Inhibits Ovarian Cancer Progression. 免疫抑制性巨噬细胞靶向LxCxE裂孔视网膜母细胞瘤蛋白抑制卵巢癌进展。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-24-0440
Evgenii N Tcyganov, Taekyoung Kwak, Xue Yang, Adi Narayana Reddy Poli, Colin Hart, Avishek Bhuniya, Joel Cassel, Andrew V Kossenkov, Noam Auslander, Lily Lu, Paridhima Sharma, Maria De Grecia Cauti Mendoza, Dmitry Zhigarev, Gwendolyn M Cramer, Mark Gregory Cadungog, Stephanie Jean, Sudeshna Chatterjee-Paer, David B Weiner, Laxminarasimha Donthireddy, Bryan Bristow, Rugang Zhang, Vladimir A Tyurin, Yulia Y Tyurina, Hülya Bayir, Valerian E Kagan, Joseph M Salvino, Luis J Montaner

Ovarian cancer remains a major health threat with limited treatment options available. It is characterized by an immunosuppressive tumor microenvironment (TME) maintained by tumor-associated macrophages (TAM), hindering antitumor responses and immunotherapy efficacy. In this study, we showed that targeting retinoblastoma protein (Rb) by disruption of its LxCxE cleft pocket caused preferential cell death in Rbhigh M2-polarized or M2-like Rbhigh immunosuppressive TAMs by induction of endoplasmic reticulum stress, p53, and mitochondria-related cell death pathways. A reduction of protumor immunosuppressive macrophages from TME in vivo resulted in enhanced T-cell infiltration and T-cell antitumor response and inhibited cancer progression. We demonstrated increased Rb expression in TAMs in women with ovarian cancer, which was associated with poorer prognosis. Ex vivo, we showed analogous cell death induction by therapeutic Rb targeting in TAMs in post-surgery ascites from patients with ovarian cancer. Overall, our data identify the therapeutic targeting of the Rb LxCxE cleft pocket as a promising approach for ovarian cancer treatment through depletion of immunosuppressive Rbhigh TAMs and re-shaping of the TME immune landscape.

卵巢癌仍然是一个主要的健康威胁,可用的治疗方案有限。其特点是肿瘤相关巨噬细胞(tam)维持免疫抑制肿瘤微环境(TME),阻碍抗肿瘤反应和免疫治疗效果。本研究表明,通过破坏视网膜母细胞瘤蛋白(Rb)的LxCxE裂袋,通过诱导内质网应激、p53和线粒体相关的细胞死亡途径,在Rbhigh M2极化或M2样Rbhigh免疫抑制性TAMs中导致优先细胞死亡。体内TME中促肿瘤免疫抑制巨噬细胞的减少增强了T细胞浸润,T细胞抗肿瘤反应,并抑制了癌症进展。我们证明,卵巢癌女性TAMs中Rb表达的增加与预后较差有关。在体外,我们在卵巢癌患者术后腹水的TAMs中发现了类似的靶向治疗性Rb诱导细胞死亡。总的来说,我们的数据阐明了靶向治疗Rb LxCxE裂袋作为一种新的有希望的卵巢癌治疗方法,通过消耗免疫抑制性Rbhigh tam和重塑TME免疫景观。
{"title":"Targeting LxCxE Cleft Pocket of Retinoblastoma Protein in Immunosuppressive Macrophages Inhibits Ovarian Cancer Progression.","authors":"Evgenii N Tcyganov, Taekyoung Kwak, Xue Yang, Adi Narayana Reddy Poli, Colin Hart, Avishek Bhuniya, Joel Cassel, Andrew V Kossenkov, Noam Auslander, Lily Lu, Paridhima Sharma, Maria De Grecia Cauti Mendoza, Dmitry Zhigarev, Gwendolyn M Cramer, Mark Gregory Cadungog, Stephanie Jean, Sudeshna Chatterjee-Paer, David B Weiner, Laxminarasimha Donthireddy, Bryan Bristow, Rugang Zhang, Vladimir A Tyurin, Yulia Y Tyurina, Hülya Bayir, Valerian E Kagan, Joseph M Salvino, Luis J Montaner","doi":"10.1158/2326-6066.CIR-24-0440","DOIUrl":"10.1158/2326-6066.CIR-24-0440","url":null,"abstract":"<p><p>Ovarian cancer remains a major health threat with limited treatment options available. It is characterized by an immunosuppressive tumor microenvironment (TME) maintained by tumor-associated macrophages (TAM), hindering antitumor responses and immunotherapy efficacy. In this study, we showed that targeting retinoblastoma protein (Rb) by disruption of its LxCxE cleft pocket caused preferential cell death in Rbhigh M2-polarized or M2-like Rbhigh immunosuppressive TAMs by induction of endoplasmic reticulum stress, p53, and mitochondria-related cell death pathways. A reduction of protumor immunosuppressive macrophages from TME in vivo resulted in enhanced T-cell infiltration and T-cell antitumor response and inhibited cancer progression. We demonstrated increased Rb expression in TAMs in women with ovarian cancer, which was associated with poorer prognosis. Ex vivo, we showed analogous cell death induction by therapeutic Rb targeting in TAMs in post-surgery ascites from patients with ovarian cancer. Overall, our data identify the therapeutic targeting of the Rb LxCxE cleft pocket as a promising approach for ovarian cancer treatment through depletion of immunosuppressive Rbhigh TAMs and re-shaping of the TME immune landscape.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1764-1782"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12532034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145022879","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
Sintilimab plus Anlotinib in Patients with Pretreated Locally Advanced or Metastatic Sarcoma: A Prospective, Single-Arm, Phase II Clinical Trial. sintilmab联合anlotinib治疗局部晚期或转移性肉瘤:一项前瞻性单组II期临床试验
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-0491
Heng Fu, Zengjun Liu, Mengyao Liu, Jing Xu, Xin Xu, Ting Hao, Guiying Wei, Hongtu Yuan, Jianbo Zhang, Dongyuan Zhu

Advanced sarcomas have limited treatment options after standard therapy, and therefore we investigated the efficacy and safety of sintilimab plus anlotinib in this setting. Patients age 18 to 75 years with advanced sarcomas and prior systemic therapy were enrolled. Patients with untreated, primary chemotherapy-resistant tumor types, such as alveolar soft-part sarcoma and clear-cell sarcoma, were also included. Patients received sintilimab 200 mg (day 1) and anlotinib (8, 10, or 12 mg investigator-chosen, day 1-14) every 3 weeks. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and adverse events (AE). The predictive value of tertiary lymphoid structure (TLS) was explored. A total of 42 patients were enrolled, and 40 (95.2%) patients were non-alveolar soft-part sarcoma. The ORR and DCR were 30.9% [95% confidence interval (CI), 16.4%-45.5%] and 76.2% (95% CI, 62.8%-89.6%), respectively, with a median follow-up duration of 15.4 months, and the median PFS was 5.0 months (95% CI, 2.8-10.2). The median OS was not reached. The most common AEs included elevated lactate dehydrogenase (28.57%), hypoproteinemia (21.43%), and increased thyroid-stimulating hormone (21.43%). The most common ≥ grade 3 AEs were hypertension (4.76%) and hyponatremia (4.76%). Two serious AEs (one hepatitis and one intestinal perforation) were recorded. The ORR in TLS-positive patients (n = 7) was significantly higher than that in TLS-negative patients (n = 28; 71.4% vs. 25.0%, P = 0.033). Therefore, sintilimab plus anlotinib demonstrated promising antitumor activity with manageable toxicity in advanced sarcomas, particularly among TLS-positive patients.

晚期肉瘤在标准治疗后的治疗选择有限,因此我们研究了辛替单抗和安洛替尼在这种情况下的疗效和安全性。年龄18-75岁的晚期肉瘤患者,既往接受过全身治疗。患者未经治疗,原发性化疗耐药肿瘤类型,如肺泡软组织肉瘤,透明细胞肉瘤等也包括在内。患者每3周接受sintilmab 200mg (d1)和anlotinib(研究者选择的8,10或12mg, d1-14)治疗。主要终点为客观缓解率(ORR)。次要终点包括无进展生存期(PFS)、总生存期(OS)、疾病控制率(DCR)和不良事件(ae)。探讨三级淋巴结构(TLS)的预测价值。共纳入42例患者,其中40例(95.2%)为非asps患者。ORR和DCR分别为30.9% (95% CI, 16.4-45.5%)和76.2% (95% CI, 62.8-89.6%),中位随访时间为15.4个月,中位PFS为5.0个月(95% CI, 2.8-10.2)。未达到中位操作系统。最常见的ae包括乳酸脱氢酶升高(28.57%)、低蛋白血症(21.43%)和促甲状腺激素升高(21.43%)。最常见的≥3级ae是高血压(4.76%)和低钠血症(4.76%)。严重不良反应2例(1例肝炎,1例肠穿孔)。tls阳性患者(n = 7)的ORR显著高于tls阴性患者(n = 28) (71.4% vs. 25.0%, P = 0.033)。因此,sintilimab + anlotinib在晚期肉瘤,特别是tls阳性患者中显示出有希望的抗肿瘤活性和可控的毒性。
{"title":"Sintilimab plus Anlotinib in Patients with Pretreated Locally Advanced or Metastatic Sarcoma: A Prospective, Single-Arm, Phase II Clinical Trial.","authors":"Heng Fu, Zengjun Liu, Mengyao Liu, Jing Xu, Xin Xu, Ting Hao, Guiying Wei, Hongtu Yuan, Jianbo Zhang, Dongyuan Zhu","doi":"10.1158/2326-6066.CIR-25-0491","DOIUrl":"10.1158/2326-6066.CIR-25-0491","url":null,"abstract":"<p><p>Advanced sarcomas have limited treatment options after standard therapy, and therefore we investigated the efficacy and safety of sintilimab plus anlotinib in this setting. Patients age 18 to 75 years with advanced sarcomas and prior systemic therapy were enrolled. Patients with untreated, primary chemotherapy-resistant tumor types, such as alveolar soft-part sarcoma and clear-cell sarcoma, were also included. Patients received sintilimab 200 mg (day 1) and anlotinib (8, 10, or 12 mg investigator-chosen, day 1-14) every 3 weeks. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and adverse events (AE). The predictive value of tertiary lymphoid structure (TLS) was explored. A total of 42 patients were enrolled, and 40 (95.2%) patients were non-alveolar soft-part sarcoma. The ORR and DCR were 30.9% [95% confidence interval (CI), 16.4%-45.5%] and 76.2% (95% CI, 62.8%-89.6%), respectively, with a median follow-up duration of 15.4 months, and the median PFS was 5.0 months (95% CI, 2.8-10.2). The median OS was not reached. The most common AEs included elevated lactate dehydrogenase (28.57%), hypoproteinemia (21.43%), and increased thyroid-stimulating hormone (21.43%). The most common ≥ grade 3 AEs were hypertension (4.76%) and hyponatremia (4.76%). Two serious AEs (one hepatitis and one intestinal perforation) were recorded. The ORR in TLS-positive patients (n = 7) was significantly higher than that in TLS-negative patients (n = 28; 71.4% vs. 25.0%, P = 0.033). Therefore, sintilimab plus anlotinib demonstrated promising antitumor activity with manageable toxicity in advanced sarcomas, particularly among TLS-positive patients.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1845-1853"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12580780/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944037","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
Tunneling CARs: Increasing CAR T-Cell Tumor Infiltration through the Overexpression of MMP-7 and Osteopontin-b. 隧道CAR:通过MMP-7和骨桥蛋白-b的过表达增加CAR - t细胞肿瘤浸润。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-25-0149
Stacey Van Pelt, Mark White, Candise Tat, Devyn Hooper, Lindsay J Talbot, Mary Kathryn McKenna, Rohan Fernandes, Cliona M Rooney, Bilal Omer

Chimeric antigen receptor (CAR) T-cell therapy has demonstrated remarkable efficacy against hematologic malignancies but has struggled to achieve comparable success in solid tumors. A key obstacle in solid tumors is the extracellular matrix (ECM), which impedes CAR T-cell infiltration. In clinical trials, neuroblastoma has shown responsiveness to GD2-directed CAR T-cell therapy; however, the failure of GD2.CAR T cells to effectively clear bulky disease-characterized by dense ECM-highlights the critical challenge of infiltration. In this study, we demonstrate that GD2.CAR T cells exhibit a unique infiltration restriction compared with other CAR T cells and endogenous T cells. A separate analysis of clinical datasets identified MMP7 and SPP1 [which encodes osteopontin (OPN)] as candidate genes to improve the infiltration of GD2.CAR T cells as these were upregulated in tumor-infiltrating leukocytes. MMP-7 and OPN overexpression enhanced CAR T-cell extravasation and interstitial movement in ECM-dense environments in vitro. Overexpression of either OPN or MMP-7 significantly improved tumor infiltration in a xenograft model of neuroblastoma. This resulted in improved tumor control and a survival extension in OPN-GD2.CAR T cell-treated mice compared with unmodified GD2.CAR T cells. OPN overexpression did not increase off-target infiltration into healthy tissues or promote tumor metastasis, highlighting its potential for safe therapeutic application. Our study provides a framework for further exploration of gene modifications to improve CAR T-cell infiltration in solid tumors and identifies OPN as a candidate to explore in this regard. See related Spotlight by Gasparetto and Chiarle, p. 1698.

嵌合抗原受体(CAR) t细胞疗法已经证明了对血液系统恶性肿瘤的显著疗效,但在实体肿瘤方面却很难取得相当的成功。实体瘤的一个关键障碍是细胞外基质(ECM),它阻碍CAR - t细胞的浸润。在临床试验中,神经母细胞瘤(NB)显示出对GD2定向CAR - t细胞治疗的反应性,然而,GD2的失败。CAR - T细胞有效清除大块疾病(以致密的ECM为特征)凸显了浸润的关键挑战。在本研究中,我们证明了GD2。与其他CAR - T细胞和内源性T细胞相比,CAR - T细胞表现出独特的浸润限制。对临床数据集的单独分析发现MMP7和SPP1(编码骨桥蛋白;OPN)是改善GD2浸润的候选基因。这些CAR - T细胞在肿瘤浸润性白细胞中表达上调。MMP-7和OPN过表达在体外ecm密集环境中增强CAR - t细胞外渗和间质运动。在NB异种移植模型中,过表达OPN或MMP-7均可显著改善肿瘤浸润。这改善了OPN-GD2的肿瘤控制,延长了生存期。与未经修饰的GD2相比。CAR - T细胞。OPN过表达不会增加健康组织的脱靶浸润或促进肿瘤转移,这突出了其安全治疗应用的潜力。我们的研究为进一步探索基因修饰以改善CAR - t细胞在实体肿瘤中的浸润提供了框架,并确定了OPN作为这方面的候选物。
{"title":"Tunneling CARs: Increasing CAR T-Cell Tumor Infiltration through the Overexpression of MMP-7 and Osteopontin-b.","authors":"Stacey Van Pelt, Mark White, Candise Tat, Devyn Hooper, Lindsay J Talbot, Mary Kathryn McKenna, Rohan Fernandes, Cliona M Rooney, Bilal Omer","doi":"10.1158/2326-6066.CIR-25-0149","DOIUrl":"10.1158/2326-6066.CIR-25-0149","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T-cell therapy has demonstrated remarkable efficacy against hematologic malignancies but has struggled to achieve comparable success in solid tumors. A key obstacle in solid tumors is the extracellular matrix (ECM), which impedes CAR T-cell infiltration. In clinical trials, neuroblastoma has shown responsiveness to GD2-directed CAR T-cell therapy; however, the failure of GD2.CAR T cells to effectively clear bulky disease-characterized by dense ECM-highlights the critical challenge of infiltration. In this study, we demonstrate that GD2.CAR T cells exhibit a unique infiltration restriction compared with other CAR T cells and endogenous T cells. A separate analysis of clinical datasets identified MMP7 and SPP1 [which encodes osteopontin (OPN)] as candidate genes to improve the infiltration of GD2.CAR T cells as these were upregulated in tumor-infiltrating leukocytes. MMP-7 and OPN overexpression enhanced CAR T-cell extravasation and interstitial movement in ECM-dense environments in vitro. Overexpression of either OPN or MMP-7 significantly improved tumor infiltration in a xenograft model of neuroblastoma. This resulted in improved tumor control and a survival extension in OPN-GD2.CAR T cell-treated mice compared with unmodified GD2.CAR T cells. OPN overexpression did not increase off-target infiltration into healthy tissues or promote tumor metastasis, highlighting its potential for safe therapeutic application. Our study provides a framework for further exploration of gene modifications to improve CAR T-cell infiltration in solid tumors and identifies OPN as a candidate to explore in this regard. See related Spotlight by Gasparetto and Chiarle, p. 1698.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1732-1748"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944048","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
期刊
Cancer immunology research
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