首页 > 最新文献

Cancer immunology research最新文献

英文 中文
TCPGdb: A comprehensive T cell perturbation genomics database for identification of critical T cell regulators. TCPGdb:一个全面的T细胞微扰基因组数据库,用于鉴定关键的T细胞调节因子。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-21 DOI: 10.1158/2326-6066.CIR-25-0168
Chuanpeng Dong, Feifei Zhang, Kaiyuan Tang, Nipun Verma, Xinxin Zhu, Di Feng, James Cai, Hongyu Zhao, Sidi Chen

Large parallel genetic screens have been used to identified targets and regulators that enhance T cell antitumor capability and persistence in tumor microenvironment. We hypothesized that by combining the pooled screen data from multiple independent genetic screens we could provide a systematic, comprehensive, and robust analysis of the effect of gene perturbation on T-cell based immunotherapies. After collecting data from previously published T cell screens, including CRISPR-based and ORF-based screens, through Gene Expression Omnibus (GEO), we reprocessed the gene hits summary and conducted a pathway enrichment analysis. A T cell screen perturbation score (TPS) metric was employed to quantify the impact of a gene perturbation on T cell function. Additionally, gene expression data (both bulk RNA level and single-cell RNA level) from autoimmune disease cohort and T cell-derived cancer patients were incorporated to gain further insight on gene perturbations that potentially augment T cell proliferation. We integrated all data and analysis on 35 T cells screens into our state-of-the-art T cell perturbation genomics database (TCPGdb), which is accessible through our webserver (http://tcpgdb.sidichenlab.org/) and allows users to interactively explore the impact of query genes on T cell function.

大型平行基因筛选已被用于识别增强T细胞抗肿瘤能力和肿瘤微环境持久性的靶标和调节因子。我们假设,通过结合来自多个独立基因筛选的汇总筛选数据,我们可以对基因扰动对t细胞免疫疗法的影响提供系统、全面和可靠的分析。通过Gene Expression Omnibus (GEO)收集先前发表的T细胞筛选(包括基于crispr和基于orf的筛选)的数据后,我们重新处理了基因命中摘要并进行了途径富集分析。采用T细胞筛选扰动评分(TPS)指标来量化基因扰动对T细胞功能的影响。此外,结合自身免疫性疾病队列和T细胞衍生性癌症患者的基因表达数据(散装RNA水平和单细胞RNA水平),以进一步了解可能增强T细胞增殖的基因扰动。我们将35个T细胞筛选的所有数据和分析整合到我们最先进的T细胞扰动基因组数据库(TCPGdb)中,该数据库可通过我们的网络服务器(http://tcpgdb.sidichenlab.org/)访问,并允许用户交互式地探索查询基因对T细胞功能的影响。
{"title":"TCPGdb: A comprehensive T cell perturbation genomics database for identification of critical T cell regulators.","authors":"Chuanpeng Dong, Feifei Zhang, Kaiyuan Tang, Nipun Verma, Xinxin Zhu, Di Feng, James Cai, Hongyu Zhao, Sidi Chen","doi":"10.1158/2326-6066.CIR-25-0168","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0168","url":null,"abstract":"<p><p>Large parallel genetic screens have been used to identified targets and regulators that enhance T cell antitumor capability and persistence in tumor microenvironment. We hypothesized that by combining the pooled screen data from multiple independent genetic screens we could provide a systematic, comprehensive, and robust analysis of the effect of gene perturbation on T-cell based immunotherapies. After collecting data from previously published T cell screens, including CRISPR-based and ORF-based screens, through Gene Expression Omnibus (GEO), we reprocessed the gene hits summary and conducted a pathway enrichment analysis. A T cell screen perturbation score (TPS) metric was employed to quantify the impact of a gene perturbation on T cell function. Additionally, gene expression data (both bulk RNA level and single-cell RNA level) from autoimmune disease cohort and T cell-derived cancer patients were incorporated to gain further insight on gene perturbations that potentially augment T cell proliferation. We integrated all data and analysis on 35 T cells screens into our state-of-the-art T cell perturbation genomics database (TCPGdb), which is accessible through our webserver (http://tcpgdb.sidichenlab.org/) and allows users to interactively explore the impact of query genes on T cell function.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145573310","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
Distinct spatially resolved tumor microenvironment trajectories define benefit from ramucirumab plus pembrolizumab in refractory PD-L1+ gastric cancer. 不同的空间分解肿瘤微环境轨迹确定了ramucirumab联合派姆单抗治疗难治性PD-L1+胃癌的益处。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-18 DOI: 10.1158/2326-6066.CIR-25-0625
Sung Hee Lim, Minae An, You Jeong Heo, Hyuk Lee, Byung-Hoon Min, Arnav Mehta, Soomin Ahn, Kyoung-Mee Kim, Seung Tae Kim, Samuel J Klempner, Jeeyun Lee

The activity of bispecific antibodies against vascular endothelial growth factor (VEGF) and programmed death ligand-1 (PD-1) have reinvigorated interest in dual VEGF and PD-1 targeting across solid tumors. However, the tumor and immune features influencing tumor response remain largely unknown. We conducted a single arm phase II trial evaluating dual VEGFR2 and PD-1 targeting with ramucirumab and pembrolizumab in pre-treated PD-L1+ gastric cancer. Twenty-six patients were enrolled between June 2021 and May 2023. Nine patients received anti-PD-1 therapy before trial enrollment. There were no complete responses, but six patients had a partial response, for an objective response rate of 23%. Median progression-free survival was 2.7 months, and median overall survival was 10.9 months. Grade ≥3 treatment-related adverse events occurred in 39% of patients. Digital spatial profiling of serial primary tumor biopsies revealed distinct tumor microenvironment phenotypes between responders and non-responders. Responders were characterized by higher baseline tumoral VEGF signaling, baseline enrichment in antigen processing and presentation in the immune compartment, shorter distances between tumor and immune cell populations, and greater on-treatment vascular normalization coupled to cytotoxic T-cell infiltration. Non-responders had greater baseline hypoxia and PDGF scores, significantly upregulated TGF-β in immune cell regions, and greater distances between tumor and immune cells and immune cells and vessels. Paired peripheral blood mass cytometry revealed lower proportions of myeloid-derived suppressor cells in responders. Together these data highlight on-treatment remodeling under the therapeutic pressure of dual VEGF and PD-1 blockade, and suggest features associated with the durable benefit seen in a subset of patients.

针对血管内皮生长因子(VEGF)和程序性死亡配体-1 (PD-1)的双特异性抗体的活性重新激发了对实体肿瘤中VEGF和PD-1双重靶向的兴趣。然而,影响肿瘤反应的肿瘤和免疫特征在很大程度上仍然未知。我们进行了一项单臂II期试验,评估ramucirumab和pembrolizumab在预处理PD-L1+胃癌中的双重VEGFR2和PD-1靶向。在2021年6月至2023年5月期间招募了26名患者。9名患者在试验入组前接受了抗pd -1治疗。没有完全缓解,但6例患者有部分缓解,客观缓解率为23%。中位无进展生存期为2.7个月,中位总生存期为10.9个月。39%的患者发生≥3级治疗相关不良事件。一系列原发肿瘤活检的数字空间分析揭示了应答者和无应答者之间不同的肿瘤微环境表型。应答者的特点是更高的基线肿瘤VEGF信号,抗原加工和免疫室递呈的基线富集,肿瘤和免疫细胞群之间的距离更短,治疗时血管正常化程度更高,加上细胞毒性t细胞浸润。无应答者的基线缺氧和PDGF评分更高,免疫细胞区TGF-β显著上调,肿瘤与免疫细胞、免疫细胞与血管之间的距离更大。配对外周血细胞计数显示应答者骨髓源性抑制细胞的比例较低。综上所述,这些数据突出了在VEGF和PD-1双重阻断的治疗压力下的治疗期重塑,并提示了在一部分患者中看到的与持久获益相关的特征。
{"title":"Distinct spatially resolved tumor microenvironment trajectories define benefit from ramucirumab plus pembrolizumab in refractory PD-L1+ gastric cancer.","authors":"Sung Hee Lim, Minae An, You Jeong Heo, Hyuk Lee, Byung-Hoon Min, Arnav Mehta, Soomin Ahn, Kyoung-Mee Kim, Seung Tae Kim, Samuel J Klempner, Jeeyun Lee","doi":"10.1158/2326-6066.CIR-25-0625","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0625","url":null,"abstract":"<p><p>The activity of bispecific antibodies against vascular endothelial growth factor (VEGF) and programmed death ligand-1 (PD-1) have reinvigorated interest in dual VEGF and PD-1 targeting across solid tumors. However, the tumor and immune features influencing tumor response remain largely unknown. We conducted a single arm phase II trial evaluating dual VEGFR2 and PD-1 targeting with ramucirumab and pembrolizumab in pre-treated PD-L1+ gastric cancer. Twenty-six patients were enrolled between June 2021 and May 2023. Nine patients received anti-PD-1 therapy before trial enrollment. There were no complete responses, but six patients had a partial response, for an objective response rate of 23%. Median progression-free survival was 2.7 months, and median overall survival was 10.9 months. Grade ≥3 treatment-related adverse events occurred in 39% of patients. Digital spatial profiling of serial primary tumor biopsies revealed distinct tumor microenvironment phenotypes between responders and non-responders. Responders were characterized by higher baseline tumoral VEGF signaling, baseline enrichment in antigen processing and presentation in the immune compartment, shorter distances between tumor and immune cell populations, and greater on-treatment vascular normalization coupled to cytotoxic T-cell infiltration. Non-responders had greater baseline hypoxia and PDGF scores, significantly upregulated TGF-β in immune cell regions, and greater distances between tumor and immune cells and immune cells and vessels. Paired peripheral blood mass cytometry revealed lower proportions of myeloid-derived suppressor cells in responders. Together these data highlight on-treatment remodeling under the therapeutic pressure of dual VEGF and PD-1 blockade, and suggest features associated with the durable benefit seen in a subset of patients.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145547929","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
The allogeneic FAP-CAR-IL15 iNKT therapy MiNK-215 remodels the tumor stroma to enhance antitumor immunity. 同种异体FAP-CAR-IL15 iNKT疗法MiNK-215重塑肿瘤基质,增强抗肿瘤免疫。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-14 DOI: 10.1158/2326-6066.CIR-25-0349
Eleni Chantzoura, Efrat Altman-Sharoni, Xavier Michelet, Martyna C Popis, Magdalena Niedzielska, Bishnu Joshi, Reed Masakayan, Gerard Rubi-Sans, Stephen Addis, Justin G Keith, Shanmugarajan Krishnan, Jin San Choi, Stephanie Sanders, Alvaro Sebastian-Yague, Shannon K Boi, Moira Pinzan-Rossi, Georgios Antonopoulos, Paul Ibbett, Deborah E Wright, Olivier Le Tonqueze, Nick Kushner, Chinar Pathak, Kah Teong Soh, Amy L Chalmers, Rachel Smith, Vignesh Venkatraman, Sara Farahi, John Pravin, Priyadarshini Iyer, Emmanuel Briend, Olga Ignatovich, Marco A Purbhoo, Tyler J Curiel, Mark A Exley, Robert B Stein, Enoch Kim, Jennifer S Buell, Marc A van Dijk, Dhan Chand

Cellular immunotherapies show remarkable efficacy against hematological malignancies. However, applying these therapies against solid tumors is challenging. Among the obstacles are the lack of tumor-specific antigens and the immunosuppressive tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) expressing fibroblast activation protein (FAP) are key contributors in shaping this immunosuppressive landscape, yet developing effective strategies for targeting these cells remains an ongoing challenge. Here, we describe the design, generation, and characterization of MiNK-215, an allogeneic human invariant natural killer T (iNKT) cell therapy in which iNKT cells were engineered to express a FAP-targeting chimeric antigen receptor (CAR) and to secrete interleukin-15 (IL-15) to remodel the TME and enhance antitumor activity. MiNK-215 modulated multifunctional immune responses by enhancing T-cell responsiveness, dendritic-cell activation, M1 macrophage polarization, and tumor killing. In a lung tumor mouse model, MiNK-215 depleted FAP+ CAFs, enhanced antigen-specific T-cell infiltration, and promoted durable antitumor immunity without off-target toxicity. These findings were extended to human organoid models of treatment-refractory Microsatellite Stable Colorectal Cancer (MSS-CRC) liver metastases, establishing FAP-CAR-IL-15 iNKT cells as a promising strategy to overcome immunotherapy resistance in solid tumors.

细胞免疫疗法对血液系统恶性肿瘤有显著疗效。然而,将这些疗法应用于实体瘤是具有挑战性的。其中的障碍是缺乏肿瘤特异性抗原和免疫抑制肿瘤微环境(TME)。表达成纤维细胞活化蛋白(FAP)的癌症相关成纤维细胞(CAFs)是形成这种免疫抑制格局的关键因素,但开发针对这些细胞的有效策略仍然是一个持续的挑战。在这里,我们描述了MiNK-215的设计,生成和表征,MiNK-215是一种异体人类不变性自然杀伤T (iNKT)细胞疗法,其中iNKT细胞被工程化表达靶向fap的嵌合抗原受体(CAR)并分泌白细胞介素-15 (IL-15)来重塑TME并增强抗肿瘤活性。MiNK-215通过增强t细胞反应性、树突状细胞活化、M1巨噬细胞极化和肿瘤杀伤来调节多功能免疫反应。在肺肿瘤小鼠模型中,MiNK-215减少FAP+ CAFs,增强抗原特异性t细胞浸润,促进持久的抗肿瘤免疫,无脱靶毒性。这些发现被扩展到治疗难治性微卫星稳定结直肠癌(MSS-CRC)肝转移的人类类器官模型,建立了FAP-CAR-IL-15 iNKT细胞作为克服实体肿瘤免疫治疗耐药的有希望的策略。
{"title":"The allogeneic FAP-CAR-IL15 iNKT therapy MiNK-215 remodels the tumor stroma to enhance antitumor immunity.","authors":"Eleni Chantzoura, Efrat Altman-Sharoni, Xavier Michelet, Martyna C Popis, Magdalena Niedzielska, Bishnu Joshi, Reed Masakayan, Gerard Rubi-Sans, Stephen Addis, Justin G Keith, Shanmugarajan Krishnan, Jin San Choi, Stephanie Sanders, Alvaro Sebastian-Yague, Shannon K Boi, Moira Pinzan-Rossi, Georgios Antonopoulos, Paul Ibbett, Deborah E Wright, Olivier Le Tonqueze, Nick Kushner, Chinar Pathak, Kah Teong Soh, Amy L Chalmers, Rachel Smith, Vignesh Venkatraman, Sara Farahi, John Pravin, Priyadarshini Iyer, Emmanuel Briend, Olga Ignatovich, Marco A Purbhoo, Tyler J Curiel, Mark A Exley, Robert B Stein, Enoch Kim, Jennifer S Buell, Marc A van Dijk, Dhan Chand","doi":"10.1158/2326-6066.CIR-25-0349","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0349","url":null,"abstract":"<p><p>Cellular immunotherapies show remarkable efficacy against hematological malignancies. However, applying these therapies against solid tumors is challenging. Among the obstacles are the lack of tumor-specific antigens and the immunosuppressive tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) expressing fibroblast activation protein (FAP) are key contributors in shaping this immunosuppressive landscape, yet developing effective strategies for targeting these cells remains an ongoing challenge. Here, we describe the design, generation, and characterization of MiNK-215, an allogeneic human invariant natural killer T (iNKT) cell therapy in which iNKT cells were engineered to express a FAP-targeting chimeric antigen receptor (CAR) and to secrete interleukin-15 (IL-15) to remodel the TME and enhance antitumor activity. MiNK-215 modulated multifunctional immune responses by enhancing T-cell responsiveness, dendritic-cell activation, M1 macrophage polarization, and tumor killing. In a lung tumor mouse model, MiNK-215 depleted FAP+ CAFs, enhanced antigen-specific T-cell infiltration, and promoted durable antitumor immunity without off-target toxicity. These findings were extended to human organoid models of treatment-refractory Microsatellite Stable Colorectal Cancer (MSS-CRC) liver metastases, establishing FAP-CAR-IL-15 iNKT cells as a promising strategy to overcome immunotherapy resistance in solid tumors.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145511775","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
CD8+ T cell antitumor immunity via human iNKT-DC conjugates. CD8+ T细胞通过人iNKT-DC偶联物抗肿瘤免疫。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-14 DOI: 10.1158/2326-6066.CIR-25-0454
Dana C Baiu, Kelsey A Smith, Sarah A Ferguson, Nathan R Schwartz, Shashwot Tripathy, Joshua Brand, Huy Q Dinh, Makoto Ohashi, Eric C Johannsen, Jenny E Gumperz

Invariant Natural Killer T (iNKT) cells are a conserved T lymphocyte population capable of acting on dendritic cells (DCs) to potently amplify downstream immune responses. However, the processes underlying such iNKT adjuvancy remain poorly understood. Here, we showed that allogeneic human CD4+ iNKT cells form stably adhered bi-cellular complexes with monocyte-derived DCs that migrated together as pairs and showed extended DC calcium signaling. Compared to DCs treated with the synthetic adjuvant monophosphoryl lipid A (MPLA), DCs complexed with iNKT cells had elevated expression of MHC class I and multiple costimulatory molecules including 4-1BBL, OX40L, and IL-15R, while the iNKT cells expressed CD70. Consistent with this distinctive co-stimulatory profile, iNKT-DC complexes were efficient activators of CD8+ T cells. Administering iNKT-DC complexes as a cellular immunotherapy in a xenograft model of aggressive human B cell lymphoma resulted in rapid reduction in tumor mass, antigen-specific B cell clearance, and transcriptional activation indicative of enhanced T cell proliferation and effector responses. iNKT-DC immunotherapy was effective at late stages of tumor progression that were refractory to immune checkpoint blockade immunotherapy, suggesting that the consortium of activating signals provided by iNKT-DC complexes rejuvenates exhausted antitumor immunity. Finally, allogeneic CD4+ iNKT cells formed similar complexes with monocyte-derived DCs from Head and Neck Cancer patients and promoted tumor antigen-dependent CD8+ T cell activation. These results show that monocyte-derived DCs paired with allogeneic CD4+ iNKT cells act as a potent antitumor cellular immunotherapy that activates antigen-specific CD8+ T cell immunity.

不变性自然杀伤T细胞(iNKT)是一种保守的T淋巴细胞群,能够作用于树突状细胞(dc),有效地放大下游免疫反应。然而,这种iNKT佐剂背后的过程仍然知之甚少。在这里,我们发现同种异体的人CD4+ iNKT细胞与单核细胞来源的DC形成稳定粘附的双细胞复合物,它们成对地一起迁移,并显示出扩展的DC钙信号。与合成佐剂单磷酰脂质A (MPLA)处理的树突细胞相比,与iNKT细胞复合的树突细胞MHC I类和4-1BBL、OX40L和IL-15R等多种共刺激分子的表达升高,而iNKT细胞表达CD70。与这种独特的共刺激特征一致,iNKT-DC复合物是CD8+ T细胞的有效激活剂。在侵袭性人B细胞淋巴瘤异种移植模型中使用iNKT-DC复合物作为细胞免疫疗法,可迅速减少肿瘤质量,抗原特异性B细胞清除,转录激活表明T细胞增殖和效应反应增强。iNKT-DC免疫治疗对免疫检查点阻断免疫治疗无效的晚期肿瘤进展有效,这表明iNKT-DC复合物提供的激活信号联合可使耗尽的抗肿瘤免疫恢复活力。最后,同种异体CD4+ iNKT细胞与头颈癌患者的单核细胞来源的dc形成类似的复合物,并促进肿瘤抗原依赖性CD8+ T细胞的活化。这些结果表明,单核细胞来源的dc与同种异体CD4+ iNKT细胞配对,可作为一种有效的抗肿瘤细胞免疫疗法,激活抗原特异性CD8+ T细胞免疫。
{"title":"CD8+ T cell antitumor immunity via human iNKT-DC conjugates.","authors":"Dana C Baiu, Kelsey A Smith, Sarah A Ferguson, Nathan R Schwartz, Shashwot Tripathy, Joshua Brand, Huy Q Dinh, Makoto Ohashi, Eric C Johannsen, Jenny E Gumperz","doi":"10.1158/2326-6066.CIR-25-0454","DOIUrl":"10.1158/2326-6066.CIR-25-0454","url":null,"abstract":"<p><p>Invariant Natural Killer T (iNKT) cells are a conserved T lymphocyte population capable of acting on dendritic cells (DCs) to potently amplify downstream immune responses. However, the processes underlying such iNKT adjuvancy remain poorly understood. Here, we showed that allogeneic human CD4+ iNKT cells form stably adhered bi-cellular complexes with monocyte-derived DCs that migrated together as pairs and showed extended DC calcium signaling. Compared to DCs treated with the synthetic adjuvant monophosphoryl lipid A (MPLA), DCs complexed with iNKT cells had elevated expression of MHC class I and multiple costimulatory molecules including 4-1BBL, OX40L, and IL-15R, while the iNKT cells expressed CD70. Consistent with this distinctive co-stimulatory profile, iNKT-DC complexes were efficient activators of CD8+ T cells. Administering iNKT-DC complexes as a cellular immunotherapy in a xenograft model of aggressive human B cell lymphoma resulted in rapid reduction in tumor mass, antigen-specific B cell clearance, and transcriptional activation indicative of enhanced T cell proliferation and effector responses. iNKT-DC immunotherapy was effective at late stages of tumor progression that were refractory to immune checkpoint blockade immunotherapy, suggesting that the consortium of activating signals provided by iNKT-DC complexes rejuvenates exhausted antitumor immunity. Finally, allogeneic CD4+ iNKT cells formed similar complexes with monocyte-derived DCs from Head and Neck Cancer patients and promoted tumor antigen-dependent CD8+ T cell activation. These results show that monocyte-derived DCs paired with allogeneic CD4+ iNKT cells act as a potent antitumor cellular immunotherapy that activates antigen-specific CD8+ T cell immunity.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12716482/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145511834","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
Pan-cancer single-cell RNA sequencing analysis refines multi-origin monocyte and macrophage lineages. 泛癌单细胞RNA测序分析细化了多起源单核细胞和巨噬细胞谱系。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-13 DOI: 10.1158/2326-6066.CIR-24-1255
Truc Do Thanh Nguyen, Andrew J Lee, Hyun Jung Park, Nameeta Shah, Bayrta Mandzhieva, Dong-Sup Lee, Inkyung Jung, Woong-Yang Park

Tumor-associated macrophages (TAMs) play crucial and important role in cancer dynamics by affecting homeostasis, immunosuppression, and angiogenesis within the tumor microenvironment. Using single-cell transcriptomics, we constructed a comprehensive atlas of myeloid cell populations across healthy and pan-cancer tissues that revealed heterogeneity. Our analysis suggested that TAMs may arise from two distinct origins: C1QC+ TAMs, which likely are derived from resident tissue macrophages, SPP1+ TAMs and ISG15+ TAMs, which appear to originate from circulating monocytes. Regarding immature myeloid-derived suppressor cells (MDSCs), we highlighted THBS1+ MDSCs and their descendants, SPP1+ TAMs, as key contributors to tumor progression, immunosuppression, and angiogenesis. We proposed a dichotomous model for TAMs, in which C1QC+ TAMs are associated with better patient outcomes, whereas the THBS1+ MDSCs - SPP1+ TAMs lineage correlates with poorer survival and unfavorable response to immunotherapy. This study offers insight into the complex interactions among monocyte-macrophage subtypes and sheds light on TAM heterogeneity and its implications for cancer progression and therapeutic strategies.

肿瘤相关巨噬细胞(tumor associated macrophages, tam)通过影响肿瘤微环境内的稳态、免疫抑制和血管生成,在肿瘤动力学中起着至关重要的作用。利用单细胞转录组学,我们构建了健康和泛癌组织中骨髓细胞群的综合图谱,揭示了异质性。我们的分析表明,tam可能有两个不同的起源:C1QC+ tam,可能来源于常驻组织巨噬细胞;SPP1+ tam和ISG15+ tam,可能来源于循环单核细胞。对于未成熟的髓源性抑制细胞(MDSCs),我们强调THBS1+ MDSCs及其后代SPP1+ tam是肿瘤进展、免疫抑制和血管生成的关键贡献者。我们提出了一个TAMs的二分模型,其中C1QC+ TAMs与更好的患者预后相关,而THBS1+ MDSCs - SPP1+ TAMs谱系与较差的生存率和对免疫治疗的不利反应相关。这项研究为单核-巨噬细胞亚型之间的复杂相互作用提供了见解,揭示了TAM的异质性及其对癌症进展和治疗策略的影响。
{"title":"Pan-cancer single-cell RNA sequencing analysis refines multi-origin monocyte and macrophage lineages.","authors":"Truc Do Thanh Nguyen, Andrew J Lee, Hyun Jung Park, Nameeta Shah, Bayrta Mandzhieva, Dong-Sup Lee, Inkyung Jung, Woong-Yang Park","doi":"10.1158/2326-6066.CIR-24-1255","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-1255","url":null,"abstract":"<p><p>Tumor-associated macrophages (TAMs) play crucial and important role in cancer dynamics by affecting homeostasis, immunosuppression, and angiogenesis within the tumor microenvironment. Using single-cell transcriptomics, we constructed a comprehensive atlas of myeloid cell populations across healthy and pan-cancer tissues that revealed heterogeneity. Our analysis suggested that TAMs may arise from two distinct origins: C1QC+ TAMs, which likely are derived from resident tissue macrophages, SPP1+ TAMs and ISG15+ TAMs, which appear to originate from circulating monocytes. Regarding immature myeloid-derived suppressor cells (MDSCs), we highlighted THBS1+ MDSCs and their descendants, SPP1+ TAMs, as key contributors to tumor progression, immunosuppression, and angiogenesis. We proposed a dichotomous model for TAMs, in which C1QC+ TAMs are associated with better patient outcomes, whereas the THBS1+ MDSCs - SPP1+ TAMs lineage correlates with poorer survival and unfavorable response to immunotherapy. This study offers insight into the complex interactions among monocyte-macrophage subtypes and sheds light on TAM heterogeneity and its implications for cancer progression and therapeutic strategies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145502089","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
T-cell Subset Features and Distributions Evolve Across the Colorectal Precancer-Cancer Spectrum. t细胞亚群特征和分布在结直肠癌前期谱系中进化。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-12 DOI: 10.1158/2326-6066.CIR-25-0481
Yasutoshi Takashima, Andressa Dias Costa, Naohiko Akimoto, Tomotaka Ugai, Phoenix Bell, Juha P Väyrynen, Jason L Hornick, Mari Mino-Kenudson, Yuxue Zhong, Satoko Ugai, Koichiro Haruki, Qian Yao, Kosuke Matsuda, Mayu Higashioka, Daniel D Buchanan, Amanda I Phipps, Ulrike Peters, Marios Giannakis, Mingyang Song, Andrew T Chan, Charles S Fuchs, Jonathan A Nowak, Shuji Ogino

The immune microenvironment is a crucial component of colorectal carcinoma (CRC) that has been well characterized, but much less is known about the immune microenvironment of CRC precursors. We hypothesized that T-cell infiltrates might differ across the colorectal neoplastic spectrum. We leveraged the prospective cohort incident-tumor biobank method, which provided formalin-fixed paraffin-embedded tumor tissue specimens (N=1,825) from 790 CRC precursors (including hyperplastic polyps, sessile serrated adenomas, traditional serrated adenomas, tubular adenomas, tubulovillous adenomas, and villous adenomas) and 1,035 CRCs. We performed an in situ multispectral immunofluorescence assay for CD3, CD4, CD8, FOXP3 (negative, low, or high expression), PTPRC (CD45RO, CD45RA), MKI67 (Ki-67), and KRT combined with supervised machine learning. CD3+CD4+ cells were more abundant than CD3+CD8+ cells in most precursors. In conventional adenomas, greater villous component correlated with fewer intraepithelial CD3+CD8+ cells. Serrated lesions, including hyperplastic polyps and sessile serrated lesions, exhibited higher densities of intraepithelial CD3+CD8+ cells compared to other precursors and carcinomas. Age strata of patients with precursors (including early-onset precursors) were not associated with differential T-cell infiltration patterns. Compared to invasive CRC, precursors generally showed higher densities of CD3+CD4+ cells and CD3+CD8+ cells with phenotypes of naive (CD45RA+CD45RO-), memory (CD45RA-CD45RO+), and regulatory (FOXP3+Low and FOXP3+High) in intraepithelial and lamina propria/stromal regions. In conclusion, T-cell infiltration patterns vary across different histopathological types of the colorectal neoplastic spectrum from precursors to invasive carcinomas. Our findings shed light on how the tumor-immune microenvironment evolves during precursor development and progression to CRC.

免疫微环境是结直肠癌(CRC)的一个重要组成部分,已被很好地表征,但对结直肠癌前体的免疫微环境知之甚少。我们假设t细胞浸润可能在结直肠肿瘤谱上有所不同。我们利用前瞻性队列事件肿瘤生物库方法,提供福尔马林固定石蜡包埋肿瘤组织标本(N=1,825),来自790例CRC前体(包括增性息肉、无柄锯齿状腺瘤、传统锯齿状腺瘤、管状腺瘤、管状绒毛状腺瘤和绒毛状腺瘤)和1,035例CRC。结合监督式机器学习,我们对CD3、CD4、CD8、FOXP3(阴性、低表达或高表达)、PTPRC (CD45RO、CD45RA)、MKI67 (Ki-67)和KRT进行了原位多光谱免疫荧光检测。在大多数前体细胞中,CD3+CD4+细胞比CD3+CD8+细胞更丰富。在常规腺瘤中,较多的绒毛成分与较少的上皮内CD3+CD8+细胞相关。锯齿状病变,包括增生性息肉和无柄锯齿状病变,与其他前体和癌相比,上皮内CD3+CD8+细胞密度更高。患有前体(包括早发性前体)的患者的年龄层与不同的t细胞浸润模式无关。与浸润性结直肠癌相比,在上皮内和固有层/间质区,前体细胞普遍表现出更高的CD3+CD4+细胞和CD3+CD8+细胞密度,表型为初始型(CD45RA+CD45RO-)、记忆型(CD45RA-CD45RO+)和调节性(FOXP3+Low和FOXP3+High)。总之,从前体到浸润性癌,t细胞浸润模式在不同的结直肠肿瘤谱的组织病理类型中有所不同。我们的研究结果揭示了肿瘤免疫微环境在前体发育和CRC进展过程中的演变。
{"title":"T-cell Subset Features and Distributions Evolve Across the Colorectal Precancer-Cancer Spectrum.","authors":"Yasutoshi Takashima, Andressa Dias Costa, Naohiko Akimoto, Tomotaka Ugai, Phoenix Bell, Juha P Väyrynen, Jason L Hornick, Mari Mino-Kenudson, Yuxue Zhong, Satoko Ugai, Koichiro Haruki, Qian Yao, Kosuke Matsuda, Mayu Higashioka, Daniel D Buchanan, Amanda I Phipps, Ulrike Peters, Marios Giannakis, Mingyang Song, Andrew T Chan, Charles S Fuchs, Jonathan A Nowak, Shuji Ogino","doi":"10.1158/2326-6066.CIR-25-0481","DOIUrl":"10.1158/2326-6066.CIR-25-0481","url":null,"abstract":"<p><p>The immune microenvironment is a crucial component of colorectal carcinoma (CRC) that has been well characterized, but much less is known about the immune microenvironment of CRC precursors. We hypothesized that T-cell infiltrates might differ across the colorectal neoplastic spectrum. We leveraged the prospective cohort incident-tumor biobank method, which provided formalin-fixed paraffin-embedded tumor tissue specimens (N=1,825) from 790 CRC precursors (including hyperplastic polyps, sessile serrated adenomas, traditional serrated adenomas, tubular adenomas, tubulovillous adenomas, and villous adenomas) and 1,035 CRCs. We performed an in situ multispectral immunofluorescence assay for CD3, CD4, CD8, FOXP3 (negative, low, or high expression), PTPRC (CD45RO, CD45RA), MKI67 (Ki-67), and KRT combined with supervised machine learning. CD3+CD4+ cells were more abundant than CD3+CD8+ cells in most precursors. In conventional adenomas, greater villous component correlated with fewer intraepithelial CD3+CD8+ cells. Serrated lesions, including hyperplastic polyps and sessile serrated lesions, exhibited higher densities of intraepithelial CD3+CD8+ cells compared to other precursors and carcinomas. Age strata of patients with precursors (including early-onset precursors) were not associated with differential T-cell infiltration patterns. Compared to invasive CRC, precursors generally showed higher densities of CD3+CD4+ cells and CD3+CD8+ cells with phenotypes of naive (CD45RA+CD45RO-), memory (CD45RA-CD45RO+), and regulatory (FOXP3+Low and FOXP3+High) in intraepithelial and lamina propria/stromal regions. In conclusion, T-cell infiltration patterns vary across different histopathological types of the colorectal neoplastic spectrum from precursors to invasive carcinomas. Our findings shed light on how the tumor-immune microenvironment evolves during precursor development and progression to CRC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12616629/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145494737","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
FAM135B Deficiency Inhibits Cytotoxic T-Cell Activity in Triple-negative Breast Cancer by Blocking the IFI16-dependent STING Pathway. FAM135B缺乏通过阻断ifi16依赖性STING通路抑制三阴性乳腺癌细胞毒性t细胞活性
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-11 DOI: 10.1158/2326-6066.CIR-25-0310
Wanmei Lin, Junze Li, Jun Wu, Bin Huang, Junguang Liang, Yuan Zhang, Liang Zhao, Guangyu Yao

Despite advances in immune checkpoint blockade (ICB) for cancer treatment, only a minority of triple-negative breast cancer (TNBC) patients derive benefits, and the underlying mechanisms remain largely unknown. Herein, sequence similarity 135 family member B (FAM135B) is identified as a regulator of antitumor immunity in TNBC. Single-cell sequencing data and functional assays demonstrate the critical role of FAM135B in activating cytotoxic T cells and improving the efficacy of ICB treatment by stimulating the STING pathway. Specifically, FAM135B interacts with IFI16, inhibiting its ubiquitination and proteasomal degradation by competitively blocking its binding to the E3 ligase TRIM21, thereby initiating the IFI16-dependent STING signaling, ultimately leading to increased cytotoxic T cell activity. The deubiquitination of IFI16 at lysine 143 and lysine 561 is crucial for FAM135B-mediated activation of the STING pathway. These findings reveal that FAM135B regulates the IFI16-dependent STING pathway and subsequent immune activation. FAM135B may represent a potential predictor of ICB therapeutic responses for TNBC patients.

尽管免疫检查点阻断(ICB)在癌症治疗方面取得了进展,但只有少数三阴性乳腺癌(TNBC)患者从中获益,其潜在机制在很大程度上仍不清楚。本文中,序列相似性135家族成员B (FAM135B)被确定为TNBC抗肿瘤免疫的调节因子。单细胞测序数据和功能分析表明FAM135B在激活细胞毒性T细胞和通过刺激STING通路提高ICB治疗效果方面发挥关键作用。具体来说,FAM135B与IFI16相互作用,通过竞争性地阻断其与E3连接酶TRIM21的结合,抑制其泛素化和蛋白酶体降解,从而启动IFI16依赖的STING信号传导,最终导致细胞毒性T细胞活性增加。IFI16在赖氨酸143和赖氨酸561处的去泛素化对于fam135b介导的STING通路激活至关重要。这些发现表明FAM135B调节ifi16依赖性STING通路和随后的免疫激活。FAM135B可能是TNBC患者ICB治疗反应的潜在预测因子。
{"title":"FAM135B Deficiency Inhibits Cytotoxic T-Cell Activity in Triple-negative Breast Cancer by Blocking the IFI16-dependent STING Pathway.","authors":"Wanmei Lin, Junze Li, Jun Wu, Bin Huang, Junguang Liang, Yuan Zhang, Liang Zhao, Guangyu Yao","doi":"10.1158/2326-6066.CIR-25-0310","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0310","url":null,"abstract":"<p><p>Despite advances in immune checkpoint blockade (ICB) for cancer treatment, only a minority of triple-negative breast cancer (TNBC) patients derive benefits, and the underlying mechanisms remain largely unknown. Herein, sequence similarity 135 family member B (FAM135B) is identified as a regulator of antitumor immunity in TNBC. Single-cell sequencing data and functional assays demonstrate the critical role of FAM135B in activating cytotoxic T cells and improving the efficacy of ICB treatment by stimulating the STING pathway. Specifically, FAM135B interacts with IFI16, inhibiting its ubiquitination and proteasomal degradation by competitively blocking its binding to the E3 ligase TRIM21, thereby initiating the IFI16-dependent STING signaling, ultimately leading to increased cytotoxic T cell activity. The deubiquitination of IFI16 at lysine 143 and lysine 561 is crucial for FAM135B-mediated activation of the STING pathway. These findings reveal that FAM135B regulates the IFI16-dependent STING pathway and subsequent immune activation. FAM135B may represent a potential predictor of ICB therapeutic responses for TNBC patients.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145494754","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
Intratumoral three-cell-type clusters are a conserved feature of endogenous antitumor immunity. 肿瘤内三细胞型簇是内源性抗肿瘤免疫的保守特征。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-07 DOI: 10.1158/2326-6066.CIR-25-0062
Sheela R Damle, Jason A Carter, Kristin E Goodsell, Jose M B Pineda, Lindsay K Dickerson, Xiuyun Jiang, Jacqueline L Mudd, Thomas Walsh, Heidi L Kenerson, Jack Cernak, Sardar Shahmir B Chauhan, Emily Beirne, Sujata Jana, Amanda L Koehne, Kiran R Vij, Marianna B Ruzinova, Raymond Yeung, Shreeram Akilesh, Eric A Collisson, Ryan C Fields, I Nicholas Crispe, Venu G Pillarisetty

Effective antitumor immunity ultimately depends on the priming and activation of tumor-specific cytotoxic CD8+ T cells; however, the role of intratumoral cell-cell immune interactions remains incompletely understood. Recent work has revealed that the temporospatial colocalization of dendritic cells (DCs), helper T cells (Th), and cytotoxic T lymphocytes (CTL) within the tumor immune microenvironment following immune checkpoint blockade correlates with clinical response. Herein, we report the integration of more than one million spatially resolved single-cell profiles across six spatial proteomic and transcriptomic assays, which demonstrated that DC:Th:CTL three-cell-type clusters were common even in immunotherapy-naïve and highly desmoplastic tumors, such as fibrolamellar carcinoma and pancreatic ductal adenocarcinoma (PDAC). We found that these immune triads were enriched for functionally important type 1 conventional DC, mature DCs enriched in immunoregulatory molecules (mregDC), CXCL13+ Th, and GZMK+ effector CTL phenotypes. Subsequent multiplex immunofluorescence imaging of more than 450 primary PDAC tumors showed that the density of antigen-presenting cell (APC):Th:CTL three-cell-type clusters was correlated with intratumoral T-cell clonal expansion and improved overall survival. These findings suggest that DC:Th:CTL triads are conserved across solid tumors and highlight the importance of intratumoral spatial niches in mediating endogenous antitumor immunity.

有效的抗肿瘤免疫最终取决于肿瘤特异性细胞毒性CD8+ T细胞的启动和激活;然而,肿瘤内细胞-细胞免疫相互作用的作用仍然不完全清楚。最近的研究表明,在免疫检查点阻断后,树突状细胞(dc)、辅助性T细胞(Th)和细胞毒性T淋巴细胞(CTL)在肿瘤免疫微环境中的时空共定位与临床反应相关。在此,我们报告了通过6个空间蛋白质组学和转录组学分析整合超过100万个空间分辨率的单细胞谱,这表明DC:Th:CTL三细胞型簇在immunotherapy-naïve和高度粘连增生的肿瘤中也很常见,如纤维板层癌和胰腺导管腺癌(PDAC)。我们发现这些免疫三联体在功能重要的1型常规DC、成熟DC中富集免疫调节分子(mregDC)、CXCL13+ Th和GZMK+效应CTL表型。随后对450多例原发性PDAC肿瘤的多重免疫荧光成像显示,抗原呈递细胞(APC):Th:CTL三细胞型簇的密度与瘤内t细胞克隆扩增和总生存率的提高有关。这些发现表明DC:Th:CTL三联体在实体肿瘤中是保守的,并强调了肿瘤内空间生态位在介导内源性抗肿瘤免疫中的重要性。
{"title":"Intratumoral three-cell-type clusters are a conserved feature of endogenous antitumor immunity.","authors":"Sheela R Damle, Jason A Carter, Kristin E Goodsell, Jose M B Pineda, Lindsay K Dickerson, Xiuyun Jiang, Jacqueline L Mudd, Thomas Walsh, Heidi L Kenerson, Jack Cernak, Sardar Shahmir B Chauhan, Emily Beirne, Sujata Jana, Amanda L Koehne, Kiran R Vij, Marianna B Ruzinova, Raymond Yeung, Shreeram Akilesh, Eric A Collisson, Ryan C Fields, I Nicholas Crispe, Venu G Pillarisetty","doi":"10.1158/2326-6066.CIR-25-0062","DOIUrl":"10.1158/2326-6066.CIR-25-0062","url":null,"abstract":"<p><p>Effective antitumor immunity ultimately depends on the priming and activation of tumor-specific cytotoxic CD8+ T cells; however, the role of intratumoral cell-cell immune interactions remains incompletely understood. Recent work has revealed that the temporospatial colocalization of dendritic cells (DCs), helper T cells (Th), and cytotoxic T lymphocytes (CTL) within the tumor immune microenvironment following immune checkpoint blockade correlates with clinical response. Herein, we report the integration of more than one million spatially resolved single-cell profiles across six spatial proteomic and transcriptomic assays, which demonstrated that DC:Th:CTL three-cell-type clusters were common even in immunotherapy-naïve and highly desmoplastic tumors, such as fibrolamellar carcinoma and pancreatic ductal adenocarcinoma (PDAC). We found that these immune triads were enriched for functionally important type 1 conventional DC, mature DCs enriched in immunoregulatory molecules (mregDC), CXCL13+ Th, and GZMK+ effector CTL phenotypes. Subsequent multiplex immunofluorescence imaging of more than 450 primary PDAC tumors showed that the density of antigen-presenting cell (APC):Th:CTL three-cell-type clusters was correlated with intratumoral T-cell clonal expansion and improved overall survival. These findings suggest that DC:Th:CTL triads are conserved across solid tumors and highlight the importance of intratumoral spatial niches in mediating endogenous antitumor immunity.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12707859/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145457607","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
RAS(ON) multi-selective inhibition drives antitumor immunity in preclinical models of NRAS-mutant melanoma. RAS(ON)多选择性抑制在nras突变黑色素瘤的临床前模型中驱动抗肿瘤免疫。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-04 DOI: 10.1158/2326-6066.CIR-25-0744
Larissa Anastacio Da Costa Carvalho, Nataliya Tovbis Shifrin, Manali S Phadke, Michael F Emmons, Ximo Pechuan-Jorge, Felix Mbuga, Oscar E Ospina, Christopher Chow, Lillian Seu, Olivia L Rose, Aparna Hegde, Nikhil I Khushalani, Elsa Quintana, Keiran S M Smalley

Targeted therapies for NRAS-mutant melanoma remain an unmet clinical need. Here, we demonstrate that RMC-7977, a preclinical RAS(ON) multi-selective inhibitor representative of the investigational agent daraxonrasib (RMC-6236), was able to elicit potent antitumor immune responses across multiple NRAS-mutant melanoma models. Treatment with RMC-7977 led to rapid tumor regressions driven by inhibition of MAPK signaling, upregulation of major histocompatibility complex (MHC) and PD-L1 proteins, and enhanced infiltration of CD4⁺ and CD8⁺ T cells. Complete responses were dependent on adaptive immunity, as both CD4⁺ and CD8⁺ T cells were essential for extended survival. Resistance to treatment was marked by reduced T-cell infiltration, loss of MHC class I expression, and expansion of myeloid-derived suppressor cells. Combining RMC-7977 with anti-PD-1 boosted cytotoxic T-cell infiltration, reprogrammed myeloid cells toward an antigen-presenting phenotype, and improved survival in models resistant to PD-1 blockade. Consistent with these preclinical data, objective clinical responses were observed in two NRAS-mutant melanoma patients treated with daraxonrasib in an ongoing Phase I/Ib clinical trial. Together, these data support the continued clinical evaluation of RAS(ON) multi-selective inhibitors for the treatment of NRAS-mutant melanoma.

靶向治疗nras突变黑色素瘤仍然是一个未满足的临床需求。在这里,我们证明了rmmc -7977,一种临床前RAS(ON)多选择性抑制剂,代表了研究药物daraxonrasib (rmmc -6236),能够在多种nras突变黑色素瘤模型中引发有效的抗肿瘤免疫反应。rmmc -7977通过抑制MAPK信号、上调主要组织相容性复合体(MHC)和PD-L1蛋白,以及增强CD4 +和CD8 + T细胞的浸润,导致肿瘤快速消退。完全应答依赖于适应性免疫,因为CD4 +和CD8 + T细胞对于延长生存都是必不可少的。对治疗的抵抗表现为t细胞浸润减少,MHC I类表达丧失,骨髓源性抑制细胞扩增。将rmmc -7977与抗PD-1联合使用可促进细胞毒性t细胞浸润,将髓细胞重编程为抗原呈递表型,并提高PD-1阻断耐药模型的存活率。与这些临床前数据一致,在一项正在进行的I/Ib期临床试验中,在两名接受daraxonrasib治疗的nras突变黑色素瘤患者中观察到客观的临床反应。总之,这些数据支持RAS(ON)多选择性抑制剂治疗nras突变黑色素瘤的持续临床评估。
{"title":"RAS(ON) multi-selective inhibition drives antitumor immunity in preclinical models of NRAS-mutant melanoma.","authors":"Larissa Anastacio Da Costa Carvalho, Nataliya Tovbis Shifrin, Manali S Phadke, Michael F Emmons, Ximo Pechuan-Jorge, Felix Mbuga, Oscar E Ospina, Christopher Chow, Lillian Seu, Olivia L Rose, Aparna Hegde, Nikhil I Khushalani, Elsa Quintana, Keiran S M Smalley","doi":"10.1158/2326-6066.CIR-25-0744","DOIUrl":"10.1158/2326-6066.CIR-25-0744","url":null,"abstract":"<p><p>Targeted therapies for NRAS-mutant melanoma remain an unmet clinical need. Here, we demonstrate that RMC-7977, a preclinical RAS(ON) multi-selective inhibitor representative of the investigational agent daraxonrasib (RMC-6236), was able to elicit potent antitumor immune responses across multiple NRAS-mutant melanoma models. Treatment with RMC-7977 led to rapid tumor regressions driven by inhibition of MAPK signaling, upregulation of major histocompatibility complex (MHC) and PD-L1 proteins, and enhanced infiltration of CD4⁺ and CD8⁺ T cells. Complete responses were dependent on adaptive immunity, as both CD4⁺ and CD8⁺ T cells were essential for extended survival. Resistance to treatment was marked by reduced T-cell infiltration, loss of MHC class I expression, and expansion of myeloid-derived suppressor cells. Combining RMC-7977 with anti-PD-1 boosted cytotoxic T-cell infiltration, reprogrammed myeloid cells toward an antigen-presenting phenotype, and improved survival in models resistant to PD-1 blockade. Consistent with these preclinical data, objective clinical responses were observed in two NRAS-mutant melanoma patients treated with daraxonrasib in an ongoing Phase I/Ib clinical trial. Together, these data support the continued clinical evaluation of RAS(ON) multi-selective inhibitors for the treatment of NRAS-mutant melanoma.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12666865/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437304","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
A Sampling of Highlights from the Literature: Article Recommendations from Our Deputy and Senior Editors. 从文献的亮点抽样:文章推荐从我们的副和高级编辑。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-11-03 DOI: 10.1158/2326-6066.CIR-13-11-WWR
{"title":"A Sampling of Highlights from the Literature: Article Recommendations from Our Deputy and Senior Editors.","authors":"","doi":"10.1158/2326-6066.CIR-13-11-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-13-11-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"13 11","pages":"1695"},"PeriodicalIF":8.2,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145430379","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