首页 > 最新文献

Journal for Immunotherapy of Cancer最新文献

英文 中文
Tumor-associated macrophages educated by IGSF9 exhibit a senescence-associated secretory phenotype to promote tumor immune escape. IGSF9诱导的肿瘤相关巨噬细胞表现出衰老相关的分泌表型,促进肿瘤免疫逃逸。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-14 DOI: 10.1136/jitc-2025-012889
Jiasheng Zhang, Xianhui Meng, Xinyu Zhao, Huiwen Luan, Fangmin Li, Ting Wang, Yuxiao Sun, Guantong Liu, Yingxin Pang, Aihui Xu, Hongying Wang, Chunling Li, Lijun Hui, Fang Li, Shuping Wei, Hong Yu, Yaopeng Wang, Zunling Li

Background: IGSF9, immunoglobulin superfamily member 9, has been reported to inhibit T cell proliferation and activation, thereby promoting tumor immune escape. Tumor-associated macrophages (TAMs), the most abundant tumor-infiltrating immune cells, play a crucial role in forming the immunosuppressive tumor microenvironment. We find that IGSF9 strongly binds to TAMs, however, how it affects TAMs function remains unreported.

Methods: The spatial transcriptomics dataset (GSE189487) was analyzed to clarify the relationship among IGSF9, senescent TAMs, and T cells. RNA-seq revealed differentially expressed genes. Flow cytometry was employed to assess the binding of IGSF9-ECD proteins to macrophages. Macrophage-educated by IGSF9 were co-cultured with T cells, and the phagocytosis was observed. Membrane-system yeast two-hybrid screening, GST pull-down, and co-IP were used to identify the binding partner of IGSF9. Cellular senescence markers and the senescence-associated secretory phenotype (SASP) were assessed by flow cytometry and immunofluorescence. LL/2-control or LL/2-Igsf9 cells were injected into C57BL/6, NSG, and monocytes-depleted C57BL/6 mice. Similarly, MC38-OVA-control or MC38-OVA-Igsf9 cells were inoculated into OT-II mice. Finally, anti-IGSF9 and its LALA-PG-mutant variant were administered to C57BL/6 mice to monitor tumor growth and SASP expression.

Results: The spatial transcriptomics dataset (GSE189487) revealed that the level of IGSF9 in tumor cells was positively correlated with the senescence of TAMs, and RNA-seq revealed the differentially expressed genes which were related to senescence. Macrophage educated by IGSF9 exhibited distinct senescence phenotypes and immunosuppressive features, and IGSF9 bound to TMUB1 to activate the IL-6/STAT3 signal pathway to trigger the above phenotype. In vivo data showed that IGSF9 could induce TAM senescence, leading to inhibition of T cell activity and formation of an immunosuppressive microenvironment, then promoting tumor immune escape. Treatment with anti-IGSF9, including the LALAPG-mutant variant that reduces antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis, mitigated TAM senescence and immunosuppression, activated T cells, and suppressed tumor growth.

Conclusions: TMUB1 as a binding protein of IGSF9 is first discovered, and TAMs educated by IGSF9 exhibit senescent and suppressive phenotype to orchestrate an inhibitory tumor microenvironment, which can be reversed by anti-IGSF9 to suppress tumor progression.

背景:IGSF9是免疫球蛋白超家族9成员,有报道称其可抑制T细胞增殖和活化,从而促进肿瘤免疫逃逸。肿瘤相关巨噬细胞(tumor- associated macrophages, tam)是最丰富的肿瘤浸润免疫细胞,在形成免疫抑制肿瘤微环境中起着至关重要的作用。我们发现IGSF9与tam强烈结合,然而,它如何影响tam的功能仍未报道。方法:分析空间转录组学数据集(GSE189487),明确IGSF9、衰老tam和T细胞之间的关系。RNA-seq显示差异表达基因。流式细胞术检测IGSF9-ECD蛋白与巨噬细胞的结合情况。将IGSF9培养的巨噬细胞与T细胞共培养,观察吞噬作用。利用膜系统酵母双杂交筛选、GST下拉和co-IP技术鉴定IGSF9的结合伙伴。采用流式细胞术和免疫荧光法检测细胞衰老标志物和衰老相关分泌表型(SASP)。将LL/2-对照或LL/2-Igsf9细胞注射到C57BL/6、NSG和单核细胞缺失的C57BL/6小鼠中。同样,将mc38 - ova对照细胞或MC38-OVA-Igsf9细胞接种于OT-II小鼠。最后,将抗igsf9及其lala - pg突变体给予C57BL/6小鼠,监测肿瘤生长和SASP表达。结果:空间转录组学数据集(GSE189487)显示肿瘤细胞中IGSF9水平与tam衰老呈正相关,RNA-seq显示与衰老相关的差异表达基因。经IGSF9教育的巨噬细胞表现出明显的衰老表型和免疫抑制特征,IGSF9结合TMUB1激活IL-6/STAT3信号通路触发上述表型。体内实验数据显示,IGSF9可诱导TAM衰老,抑制T细胞活性,形成免疫抑制微环境,促进肿瘤免疫逃逸。抗igsf9治疗,包括lalapg突变变体,可降低抗体依赖性细胞毒性和抗体依赖性细胞吞噬,减轻TAM衰老和免疫抑制,激活T细胞,抑制肿瘤生长。结论:TMUB1作为IGSF9的结合蛋白首次被发现,IGSF9教育的tam表现出衰老和抑制表型,协调抑制肿瘤微环境,可通过抗IGSF9逆转,抑制肿瘤进展。
{"title":"Tumor-associated macrophages educated by IGSF9 exhibit a senescence-associated secretory phenotype to promote tumor immune escape.","authors":"Jiasheng Zhang, Xianhui Meng, Xinyu Zhao, Huiwen Luan, Fangmin Li, Ting Wang, Yuxiao Sun, Guantong Liu, Yingxin Pang, Aihui Xu, Hongying Wang, Chunling Li, Lijun Hui, Fang Li, Shuping Wei, Hong Yu, Yaopeng Wang, Zunling Li","doi":"10.1136/jitc-2025-012889","DOIUrl":"https://doi.org/10.1136/jitc-2025-012889","url":null,"abstract":"<p><strong>Background: </strong>IGSF9, immunoglobulin superfamily member 9, has been reported to inhibit T cell proliferation and activation, thereby promoting tumor immune escape. Tumor-associated macrophages (TAMs), the most abundant tumor-infiltrating immune cells, play a crucial role in forming the immunosuppressive tumor microenvironment. We find that IGSF9 strongly binds to TAMs, however, how it affects TAMs function remains unreported.</p><p><strong>Methods: </strong>The spatial transcriptomics dataset (GSE189487) was analyzed to clarify the relationship among IGSF9, senescent TAMs, and T cells. RNA-seq revealed differentially expressed genes. Flow cytometry was employed to assess the binding of IGSF9-ECD proteins to macrophages. Macrophage-educated by IGSF9 were co-cultured with T cells, and the phagocytosis was observed. Membrane-system yeast two-hybrid screening, GST pull-down, and co-IP were used to identify the binding partner of IGSF9. Cellular senescence markers and the senescence-associated secretory phenotype (SASP) were assessed by flow cytometry and immunofluorescence. LL/2-control or LL/2-<i>Igsf9</i> cells were injected into C57BL/6, NSG, and monocytes-depleted C57BL/6 mice. Similarly, MC38-<i>OVA</i>-control or <i>MC38-OVA-Igsf9</i> cells were inoculated into OT-II mice. Finally, anti-IGSF9 and its LALA-PG-mutant variant were administered to C57BL/6 mice to monitor tumor growth and SASP expression.</p><p><strong>Results: </strong>The spatial transcriptomics dataset (GSE189487) revealed that the level of IGSF9 in tumor cells was positively correlated with the senescence of TAMs, and RNA-seq revealed the differentially expressed genes which were related to senescence. Macrophage educated by IGSF9 exhibited distinct senescence phenotypes and immunosuppressive features, and IGSF9 bound to TMUB1 to activate the IL-6/STAT3 signal pathway to trigger the above phenotype. In vivo data showed that IGSF9 could induce TAM senescence, leading to inhibition of T cell activity and formation of an immunosuppressive microenvironment, then promoting tumor immune escape. Treatment with anti-IGSF9, including the LALAPG-mutant variant that reduces antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis, mitigated TAM senescence and immunosuppression, activated T cells, and suppressed tumor growth.</p><p><strong>Conclusions: </strong>TMUB1 as a binding protein of IGSF9 is first discovered, and TAMs educated by IGSF9 exhibit senescent and suppressive phenotype to orchestrate an inhibitory tumor microenvironment, which can be reversed by anti-IGSF9 to suppress tumor progression.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145985008","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
Cadonilimab (mono anti-PD-1/CTLA-4 therapy) in advanced esophageal squamous cell carcinoma. 卡多尼单抗治疗晚期食管鳞状细胞癌。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-14 DOI: 10.1136/jitc-2025-014391
Bi-Cheng Wang
{"title":"Cadonilimab (mono anti-PD-1/CTLA-4 therapy) in advanced esophageal squamous cell carcinoma.","authors":"Bi-Cheng Wang","doi":"10.1136/jitc-2025-014391","DOIUrl":"https://doi.org/10.1136/jitc-2025-014391","url":null,"abstract":"","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145984954","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
Neoadjuvant lenvatinib plus pembrolizumab in Merkel cell carcinoma: an investigator-initiated, open-label phase II trial. 新辅助lenvatinib + pembrolizumab治疗Merkel细胞癌:一项研究者发起的开放标签II期试验
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-14 DOI: 10.1136/jitc-2025-013939
Andrew S Brohl, Vernon K Sondak, Evan J Wuthrick, Younchul Kim, Zeynep Eroglu, Joseph Markowitz, Ahmad A Tarhini, Wenyi Fan, Justin Martin, Lymon Sneed, Matthew C Perez, Amod Sarnaik, Michael Harrington, Rogerio I Neves, Ricardo J Gonzalez, C Wayne Cruse, Jonathan S Zager, Kenneth Y Tsai, Nikhil I Khushalani

Background: Given the success of checkpoint inhibitor therapy in the advanced Merkel cell carcinoma (MCC) setting, there is interest in exploring immunotherapy as a neoadjuvant approach. We report the primary results of a neoadjuvant study of lenvatinib plus pembrolizumab in resectable MCC.

Methods: In this single-center, phase II open-label trial, resectable stage II-IV MCC patients received 6 weeks of neoadjuvant therapy with lenvatinib 20 mg orally daily plus pembrolizumab 200 mg intravenous dose every 3 weeks. Following local therapy, patients received continued adjuvant pembrolizumab monotherapy to complete a total treatment duration of 1 year. Pathological complete response (pCR) rate was the primary endpoint of the study.

Results: 26 patients were enrolled, including 5 (19.2%) with clinical stage II disease, 20 (76.9%) with stage III, and 1 (3.8%) with stage IV. Following neoadjuvant treatment, 2 patients (7.7%) were unable to undergo planned surgery, one due to progressive disease and one due to toxicity. On intention to treat, 15 of the 26 patients (57.7%) achieved pCR. Among 22 radiographically evaluable patients, 16 (72.7%) achieved an objective response. At a median follow-up of 20.0 months, median progression-free survival (PFS) has not been reached. PFS significantly correlated with radiographic response to neoadjuvant therapy. pCR was associated with superior PFS, though this result was not statistically significant (p=0.22). Grade 3 treatment-related adverse events (TRAEs) occurred in 14 patients (53.8%), most commonly grade 3 hypertension in 11 patients (42.3%). No grade 4-5 TRAEs were observed.

Conclusions: Lenvatinib plus pembrolizumab demonstrated encouraging efficacy with anticipated toxicity when used as neoadjuvant therapy for MCC. Further investigation of these promising findings is warranted.

Trial registration number: NCT04869137.

背景:鉴于检查点抑制剂治疗晚期默克尔细胞癌(MCC)的成功,人们有兴趣探索免疫治疗作为一种新辅助方法。我们报告了lenvatinib + pembrolizumab治疗可切除MCC的新辅助研究的主要结果。方法:在这项单中心II期开放标签试验中,可切除的II- iv期MCC患者接受6周的新辅助治疗,每日口服lenvatinib 20mg,每3周静脉注射派姆单抗200mg。局部治疗后,患者继续接受辅助派姆单抗单药治疗,完成总治疗时间为1年。病理完全缓解(pCR)率是研究的主要终点。结果:纳入26例患者,其中临床II期5例(19.2%),III期20例(76.9%),IV期1例(3.8%)。经新辅助治疗后,2例(7.7%)患者无法按计划手术,1例因病情进展,1例因毒性。在意向治疗方面,26例患者中有15例(57.7%)达到pCR。在22例放射学可评估的患者中,16例(72.7%)达到客观反应。在中位随访20.0个月时,中位无进展生存期(PFS)尚未达到。PFS与新辅助治疗的放射学反应显著相关。pCR与更好的PFS相关,尽管这个结果没有统计学意义(p=0.22)。3级治疗相关不良事件(TRAEs)发生在14例(53.8%)患者中,最常见的3级高血压发生在11例(42.3%)患者中。未见4 ~ 5级trae。结论:Lenvatinib + pembrolizumab作为MCC的新辅助治疗显示出令人鼓舞的疗效和预期的毒性。有必要对这些有希望的发现进行进一步调查。试验注册号:NCT04869137。
{"title":"Neoadjuvant lenvatinib plus pembrolizumab in Merkel cell carcinoma: an investigator-initiated, open-label phase II trial.","authors":"Andrew S Brohl, Vernon K Sondak, Evan J Wuthrick, Younchul Kim, Zeynep Eroglu, Joseph Markowitz, Ahmad A Tarhini, Wenyi Fan, Justin Martin, Lymon Sneed, Matthew C Perez, Amod Sarnaik, Michael Harrington, Rogerio I Neves, Ricardo J Gonzalez, C Wayne Cruse, Jonathan S Zager, Kenneth Y Tsai, Nikhil I Khushalani","doi":"10.1136/jitc-2025-013939","DOIUrl":"https://doi.org/10.1136/jitc-2025-013939","url":null,"abstract":"<p><strong>Background: </strong>Given the success of checkpoint inhibitor therapy in the advanced Merkel cell carcinoma (MCC) setting, there is interest in exploring immunotherapy as a neoadjuvant approach. We report the primary results of a neoadjuvant study of lenvatinib plus pembrolizumab in resectable MCC.</p><p><strong>Methods: </strong>In this single-center, phase II open-label trial, resectable stage II-IV MCC patients received 6 weeks of neoadjuvant therapy with lenvatinib 20 mg orally daily plus pembrolizumab 200 mg intravenous dose every 3 weeks. Following local therapy, patients received continued adjuvant pembrolizumab monotherapy to complete a total treatment duration of 1 year. Pathological complete response (pCR) rate was the primary endpoint of the study.</p><p><strong>Results: </strong>26 patients were enrolled, including 5 (19.2%) with clinical stage II disease, 20 (76.9%) with stage III, and 1 (3.8%) with stage IV. Following neoadjuvant treatment, 2 patients (7.7%) were unable to undergo planned surgery, one due to progressive disease and one due to toxicity. On intention to treat, 15 of the 26 patients (57.7%) achieved pCR. Among 22 radiographically evaluable patients, 16 (72.7%) achieved an objective response. At a median follow-up of 20.0 months, median progression-free survival (PFS) has not been reached. PFS significantly correlated with radiographic response to neoadjuvant therapy. pCR was associated with superior PFS, though this result was not statistically significant (p=0.22). Grade 3 treatment-related adverse events (TRAEs) occurred in 14 patients (53.8%), most commonly grade 3 hypertension in 11 patients (42.3%). No grade 4-5 TRAEs were observed.</p><p><strong>Conclusions: </strong>Lenvatinib plus pembrolizumab demonstrated encouraging efficacy with anticipated toxicity when used as neoadjuvant therapy for MCC. Further investigation of these promising findings is warranted.</p><p><strong>Trial registration number: </strong>NCT04869137.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145985011","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
CD47 destabilization via manipulating the SPOP-USP2 axis augments macrophage phagocytosis and cancer immunotherapy. 通过操纵SPOP-USP2轴来破坏CD47的稳定,增强了巨噬细胞吞噬和癌症免疫治疗。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-14 DOI: 10.1136/jitc-2025-013498
Peiqiang Yan, Xia Bu, Tao Hou, Li Chen, Guoxuan Zhong, Daoyuan Huang, Jingchao Wang, Yihang Qi, Weiwei Jiang, Zhe Li, Xutong Xue, Yang Gao, Jing Liu, Hiroyuki Inuzuka, Gordon J Freeman, Wenyi Wei, Xiaoming Dai

Background: Macrophages can eliminate cancer cells through phagocytosis via the CD47/signal regulatory protein α axis, which provides promising targets for cancer immunotherapy as innate immune checkpoints. Although CD47 is overexpressed in multiple cancer types, it remains largely unknown whether and how CD47 can be targeted by manipulating its protein stability.

Experimental design: Multiple human cancer cell lines were used to identify the function of the ubiquitin-specific protease 2 (USP2) /speckle-type POZ protein (SPOP) axis and the USP2 inhibitor on CD47 protein stability by immunoblot and immunoprecipitation, real-time quantitative PCR, in vitro deubiquitination assay, cell fractionation assay, flow cytometry, and phagocytosis assay. We investigated the antitumor immune response and immunotherapy effects of the USP2 inhibitor using multiple syngeneic and orthotopic mouse tumor models, bioluminescence imaging, immune cell depletion, tumor-infiltrating lymphocyte (TIL) isolation, and flow cytometry.

Results: Here, we report that ML364, an inhibitor of the USP2 deubiquitinase, reduces the protein abundance of CD47. Mechanistically, USP2 deubiquitinates and protects CD47 from proteasome-mediated degradation. Furthermore, we reveal that USP2 itself can be ubiquitinated by the SPOP ubiquitin E3 ligase, which leads to USP2 degradation and decreased CD47 protein abundance. Functionally, ML364 promotes macrophage phagocytosis of cancer cells by reducing the expression of CD47 and enhances the efficacy of anti-programmed cell death protein-1 (PD-1) immunotherapy, thereby inhibiting tumor growth and improving the overall survival rate in multiple syngeneic and orthotopic mouse tumor models. Bioinformatic analyses indicate that low USP2 expression or high SPOP expression predicts a better response to anti-PD-1 treatment.

Conclusion: Hence, our findings reveal a pivotal role of the SPOP/USP2 axis in regulating CD47 protein stability and advocate for combining USP2 inhibitors with anti-PD-1 immunotherapy to combat cancer.

背景:巨噬细胞可以通过CD47/信号调节蛋白α轴吞噬癌细胞,作为先天免疫检查点为肿瘤免疫治疗提供了有希望的靶点。尽管CD47在多种癌症类型中过度表达,但CD47是否以及如何通过操纵其蛋白质稳定性来靶向仍然是未知的。实验设计:采用免疫印迹、免疫沉淀、实时定量PCR、体外去泛素化实验、细胞分离实验、流式细胞术、吞噬实验等方法,研究多种人癌细胞系泛素特异性蛋白酶2 (USP2) /斑点型POZ蛋白(SPOP)轴及USP2抑制剂对CD47蛋白稳定性的影响。我们采用多种同基因和原位小鼠肿瘤模型、生物发光成像、免疫细胞消耗、肿瘤浸润淋巴细胞(TIL)分离和流式细胞术研究了USP2抑制剂的抗肿瘤免疫应答和免疫治疗效果。结果:在这里,我们报道了ML364, USP2去泛素酶的抑制剂,降低CD47的蛋白丰度。在机制上,USP2去泛素化并保护CD47免受蛋白酶体介导的降解。此外,我们发现USP2本身可以被SPOP泛素E3连接酶泛素化,从而导致USP2降解和CD47蛋白丰度降低。在功能上,ML364通过降低CD47的表达,促进癌细胞的巨噬细胞吞噬,增强抗程序性细胞死亡蛋白-1 (anti-programmed cell death protein-1, PD-1)免疫治疗的效果,从而抑制肿瘤生长,提高多种同基因和原位小鼠肿瘤模型的总生存率。生物信息学分析表明,低USP2表达或高SPOP表达预示着抗pd -1治疗的更好反应。结论:因此,我们的研究结果揭示了SPOP/USP2轴在调节CD47蛋白稳定性中的关键作用,并提倡将USP2抑制剂与抗pd -1免疫疗法联合使用以对抗癌症。
{"title":"CD47 destabilization via manipulating the SPOP-USP2 axis augments macrophage phagocytosis and cancer immunotherapy.","authors":"Peiqiang Yan, Xia Bu, Tao Hou, Li Chen, Guoxuan Zhong, Daoyuan Huang, Jingchao Wang, Yihang Qi, Weiwei Jiang, Zhe Li, Xutong Xue, Yang Gao, Jing Liu, Hiroyuki Inuzuka, Gordon J Freeman, Wenyi Wei, Xiaoming Dai","doi":"10.1136/jitc-2025-013498","DOIUrl":"https://doi.org/10.1136/jitc-2025-013498","url":null,"abstract":"<p><strong>Background: </strong>Macrophages can eliminate cancer cells through phagocytosis via the CD47/signal regulatory protein α axis, which provides promising targets for cancer immunotherapy as innate immune checkpoints. Although CD47 is overexpressed in multiple cancer types, it remains largely unknown whether and how CD47 can be targeted by manipulating its protein stability.</p><p><strong>Experimental design: </strong>Multiple human cancer cell lines were used to identify the function of the ubiquitin-specific protease 2 (USP2) /speckle-type POZ protein (SPOP) axis and the USP2 inhibitor on CD47 protein stability by immunoblot and immunoprecipitation, real-time quantitative PCR, in vitro deubiquitination assay, cell fractionation assay, flow cytometry, and phagocytosis assay. We investigated the antitumor immune response and immunotherapy effects of the USP2 inhibitor using multiple syngeneic and orthotopic mouse tumor models, bioluminescence imaging, immune cell depletion, tumor-infiltrating lymphocyte (TIL) isolation, and flow cytometry.</p><p><strong>Results: </strong>Here, we report that ML364, an inhibitor of the USP2 deubiquitinase, reduces the protein abundance of CD47. Mechanistically, USP2 deubiquitinates and protects CD47 from proteasome-mediated degradation. Furthermore, we reveal that USP2 itself can be ubiquitinated by the SPOP ubiquitin E3 ligase, which leads to USP2 degradation and decreased CD47 protein abundance. Functionally, ML364 promotes macrophage phagocytosis of cancer cells by reducing the expression of CD47 and enhances the efficacy of anti-programmed cell death protein-1 (PD-1) immunotherapy, thereby inhibiting tumor growth and improving the overall survival rate in multiple syngeneic and orthotopic mouse tumor models. Bioinformatic analyses indicate that low USP2 expression or high SPOP expression predicts a better response to anti-PD-1 treatment.</p><p><strong>Conclusion: </strong>Hence, our findings reveal a pivotal role of the SPOP/USP2 axis in regulating CD47 protein stability and advocate for combining USP2 inhibitors with anti-PD-1 immunotherapy to combat cancer.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145984960","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
Trispecific targeting of T cells engineered with TCR mimic antibodies to limit antigen escape. 用TCR模拟抗体工程化的三特异性靶向T细胞以限制抗原逃逸。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-14 DOI: 10.1136/jitc-2025-013685
Tao Dao, Guangyan Xiong, Jeremy Meyerberg, Zita Aretz, Akihiko Shiiya, Tatyana Korontsvit, Jingbao Liu, Ziyou Cui, Neel Panchwagh, Winson Cai, Chenyang Zhan, Hongbing Zhang, Cheng Liu, David A Scheinberg

Background: Antigen loss and tumor heterogeneity present significant challenges for successful immunotherapies. T-cell receptor (TCR)-based therapies rely on the recognition of epitopes derived from intracellular tumor proteins presented by major histocompatibility complex class I molecules on cell surface. Solid tumor cells frequently lack immunoproteasomes, which are crucial for processing and presenting certain immunogenic epitopes. An effective strategy to mitigate the risk of antigen absence and tumor heterogeneity is to simultaneously target multiple tumor antigens, thereby providing critical rescue from disease relapse. Previously, we engineered a TCR mimic monoclonal antibody (TCRm) "ESK2", specific for Wilm's tumor 1 (WT1)-derived epitope RMFPNAPYL (RMF) in the context of HLA-A2, into a new chimeric antigen receptor T-cell format, antibody-TCR receptor (AbTCR)-chimeric signaling receptor (CSR). However, the RMF epitope is largely dependent on processing by the immunoproteasomes, which can be lost from leukemia cells and sometimes absent in solid tumor cells.

Methods: To mitigate antigen loss, tumor heterogeneity and broaden the reach of AbTCR T cells, we combined ESK2 with a new TCRm for an immunoproteosome-independent epitope derived from WT1, VLDFAPPGA (VLD), in the context of HLA-A2 molecules, named ESK3. ESK2 and ESK3 were tandemly engineered into one AbTCR-CSR construct, simultaneously recognizing both the WT1 RMF and VLD epitopes. To add additional specificity and potency, a CSR in these cells was engineered with a single chain variable fragment (scFv) for either CD33 to treat leukemia or mesothelin to treat solid tumors. The specificity and efficacy of the AbTCR-CSRs were evaluated in both in vitro and in vivo.

Results: In vitro studies demonstrated that the Tri-AbTCR-CSR (CD33 CSR) T cells showed the best killing activity against most acute myeloid leukemia cells. Similar levels of cytotoxicity were exhibited by ESK3 AbTCR-CSR (mesothelin CSR) against most solid tumor cell lines when compared with the Tri-AbTCR or a combination of ESK2 and ESK3 AbTCR-CSR. In animal therapy models, trispecific AbTCR-CSR T cells showed efficacy equivalent to single ESK2-AbTCR or ESK3-AbTCR-CSR T cells, against hematopoietic or solid tumor cells, further supporting the advantage of triple targeting strategy, overcoming epitope loss variants.

Conclusions: Trispecific T cells targeting immunoproteasome-dependent and independent epitopes of WT1 peptide/HLA-A2 complexes, plus a CSR recognizing a third tumor-associated antigen, present an effective and cost-efficient approach for overcoming tumor immune evasion.

背景:抗原丢失和肿瘤异质性对成功的免疫治疗提出了重大挑战。基于t细胞受体(TCR)的治疗依赖于对细胞内肿瘤蛋白衍生的表位的识别,这些蛋白由细胞表面的主要组织相容性复合体I类分子呈现。实体肿瘤细胞经常缺乏免疫蛋白酶体,这对于加工和呈现某些免疫原性表位至关重要。降低抗原缺失和肿瘤异质性风险的有效策略是同时靶向多种肿瘤抗原,从而为疾病复发提供关键的拯救。之前,我们设计了一种TCR模拟单克隆抗体(TCRm)“ESK2”,在HLA-A2背景下特异性针对Wilm's tumor 1 (WT1)衍生的表位RMFPNAPYL (RMF),形成一种新的嵌合抗原受体t细胞格式,抗体-TCR受体(AbTCR)-嵌合信号受体(CSR)。然而,RMF表位在很大程度上依赖于免疫蛋白酶体的加工,这可能在白血病细胞中丢失,有时在实体瘤细胞中不存在。方法:为了减轻抗原丢失、肿瘤异质性和扩大AbTCR T细胞的作用范围,我们将ESK2与一种新的TCRm结合起来,在HLA-A2分子的背景下,将来自WT1的免疫蛋白体独立表位VLDFAPPGA (VLD)命名为ESK3。ESK2和ESK3被串联成一个AbTCR-CSR构建体,同时识别WT1 RMF和VLD表位。为了增加额外的特异性和效力,这些细胞中的CSR被设计为单链可变片段(scFv),用于CD33治疗白血病或间皮素治疗实体瘤。在体外和体内评价AbTCR-CSRs的特异性和有效性。结果:体外研究表明,Tri-AbTCR-CSR (CD33 CSR) T细胞对大多数急性髓系白血病细胞表现出最好的杀伤活性。与Tri-AbTCR或ESK2和ESK3 AbTCR-CSR的组合相比,ESK3 AbTCR-CSR(间皮素CSR)对大多数实体肿瘤细胞系表现出相似水平的细胞毒性。在动物治疗模型中,三特异性AbTCR-CSR T细胞对造血细胞或实体肿瘤细胞的疗效相当于单个ESK2-AbTCR或ESK3-AbTCR-CSR T细胞,进一步支持三重靶向策略的优势,克服表位丢失变异。结论:靶向免疫蛋白酶体依赖和独立的WT1肽/HLA-A2复合物表位的三特异性T细胞,加上识别第三种肿瘤相关抗原的CSR,为克服肿瘤免疫逃避提供了一种有效且经济的方法。
{"title":"Trispecific targeting of T cells engineered with TCR mimic antibodies to limit antigen escape.","authors":"Tao Dao, Guangyan Xiong, Jeremy Meyerberg, Zita Aretz, Akihiko Shiiya, Tatyana Korontsvit, Jingbao Liu, Ziyou Cui, Neel Panchwagh, Winson Cai, Chenyang Zhan, Hongbing Zhang, Cheng Liu, David A Scheinberg","doi":"10.1136/jitc-2025-013685","DOIUrl":"https://doi.org/10.1136/jitc-2025-013685","url":null,"abstract":"<p><strong>Background: </strong>Antigen loss and tumor heterogeneity present significant challenges for successful immunotherapies. T-cell receptor (TCR)-based therapies rely on the recognition of epitopes derived from intracellular tumor proteins presented by major histocompatibility complex class I molecules on cell surface. Solid tumor cells frequently lack immunoproteasomes, which are crucial for processing and presenting certain immunogenic epitopes. An effective strategy to mitigate the risk of antigen absence and tumor heterogeneity is to simultaneously target multiple tumor antigens, thereby providing critical rescue from disease relapse. Previously, we engineered a TCR mimic monoclonal antibody (TCRm) \"ESK2\", specific for Wilm's tumor 1 (WT1)-derived epitope RMFPNAPYL (RMF) in the context of HLA-A2, into a new chimeric antigen receptor T-cell format, antibody-TCR receptor (AbTCR)-chimeric signaling receptor (CSR). However, the RMF epitope is largely dependent on processing by the immunoproteasomes, which can be lost from leukemia cells and sometimes absent in solid tumor cells.</p><p><strong>Methods: </strong>To mitigate antigen loss, tumor heterogeneity and broaden the reach of AbTCR T cells, we combined ESK2 with a new TCRm for an immunoproteosome-independent epitope derived from WT1, VLDFAPPGA (VLD), in the context of HLA-A2 molecules, named ESK3. ESK2 and ESK3 were tandemly engineered into one AbTCR-CSR construct, simultaneously recognizing both the WT1 RMF and VLD epitopes. To add additional specificity and potency, a CSR in these cells was engineered with a single chain variable fragment (scFv) for either CD33 to treat leukemia or mesothelin to treat solid tumors. The specificity and efficacy of the AbTCR-CSRs were evaluated in both in vitro and in vivo.</p><p><strong>Results: </strong>In vitro studies demonstrated that the Tri-AbTCR-CSR (CD33 CSR) T cells showed the best killing activity against most acute myeloid leukemia cells. Similar levels of cytotoxicity were exhibited by ESK3 AbTCR-CSR (mesothelin CSR) against most solid tumor cell lines when compared with the Tri-AbTCR or a combination of ESK2 and ESK3 AbTCR-CSR. In animal therapy models, trispecific AbTCR-CSR T cells showed efficacy equivalent to single ESK2-AbTCR or ESK3-AbTCR-CSR T cells, against hematopoietic or solid tumor cells, further supporting the advantage of triple targeting strategy, overcoming epitope loss variants.</p><p><strong>Conclusions: </strong>Trispecific T cells targeting immunoproteasome-dependent and independent epitopes of WT1 peptide/HLA-A2 complexes, plus a CSR recognizing a third tumor-associated antigen, present an effective and cost-efficient approach for overcoming tumor immune evasion.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145984925","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
Merkel cell carcinoma immunotherapy: key questions in the era of immune checkpoint blockade. 默克尔细胞癌免疫治疗:免疫检查点阻断时代的关键问题。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-13 DOI: 10.1136/jitc-2025-014090
Jürgen C Becker, Georg Lodde, Maximilian Haist, Selma Ugurel

Merkel cell carcinoma (MCC) exemplifies the paradigm of immunogenic tumors that nonetheless develop sophisticated immune evasion mechanisms. This is in line with the observation that MCC exhibits remarkable susceptibility to immune checkpoint inhibitors (ICIs), with responses in approximately half of patients with advanced disease in the first-line setting. However, 40-50% of patients show primary ICI resistance, while 20-30% of patients develop acquired ICI resistance on initial disease control. Still, the advent of ICI therapy represents a revolutionary rather than evolutionary advance in MCC management, fundamentally transforming outcomes from the dismal prognosis associated with conventional chemotherapy to durable responses extending substantially beyond 2 years. Still, important questions remain: (1) Is programmed cell death protein-1 or programmed death-ligand 1 inhibition more effective? (2) How long should ICI treatment continue? (3) What is the best choice of salvage therapy for patients with primary or acquired ICI resistance? (4) Which combination regimens can increase the share of patients benefiting from ICI? (5) Which patient/tumor characteristics predict response and its duration? Finally, (6) which timing of ICI therapy, that is, the neoadjuvant, adjuvant or therapeutic setting, offers the optimal overall outcomes for patients with MCC? A number of recent studies provide initial, but unfortunately not yet definitive answers.

默克尔细胞癌(MCC)是免疫原性肿瘤的典型例子,尽管如此,它仍发展出复杂的免疫逃避机制。这与观察到的MCC对免疫检查点抑制剂(ICIs)表现出显着的易感性一致,在一线环境中,大约一半的晚期疾病患者有反应。然而,40-50%的患者表现为原发性ICI耐药,而20-30%的患者在最初的疾病控制中出现获得性ICI耐药。尽管如此,ICI治疗的出现代表了MCC管理的革命性进步,而不是革命性的进步,从根本上改变了与传统化疗相关的预后不良的结果,使其持久的反应大大超过2年。然而,重要的问题仍然存在:(1)程序性细胞死亡蛋白1或程序性死亡配体1抑制更有效?(2) ICI治疗应持续多长时间?(3)对于原发或获得性ICI耐药的患者,抢救治疗的最佳选择是什么?(4)哪种联合方案可以增加ICI患者受益的比例?(5)哪些患者/肿瘤特征可以预测反应及其持续时间?最后,(6)ICI治疗的哪个时机,即新辅助,辅助或治疗设置,为MCC患者提供最佳的总体结果?最近的一些研究提供了初步的,但不幸的是还没有确定的答案。
{"title":"Merkel cell carcinoma immunotherapy: key questions in the era of immune checkpoint blockade.","authors":"Jürgen C Becker, Georg Lodde, Maximilian Haist, Selma Ugurel","doi":"10.1136/jitc-2025-014090","DOIUrl":"https://doi.org/10.1136/jitc-2025-014090","url":null,"abstract":"<p><p>Merkel cell carcinoma (MCC) exemplifies the paradigm of immunogenic tumors that nonetheless develop sophisticated immune evasion mechanisms. This is in line with the observation that MCC exhibits remarkable susceptibility to immune checkpoint inhibitors (ICIs), with responses in approximately half of patients with advanced disease in the first-line setting. However, 40-50% of patients show primary ICI resistance, while 20-30% of patients develop acquired ICI resistance on initial disease control. Still, the advent of ICI therapy represents a revolutionary rather than evolutionary advance in MCC management, fundamentally transforming outcomes from the dismal prognosis associated with conventional chemotherapy to durable responses extending substantially beyond 2 years. Still, important questions remain: (1) Is programmed cell death protein-1 or programmed death-ligand 1 inhibition more effective? (2) How long should ICI treatment continue? (3) What is the best choice of salvage therapy for patients with primary or acquired ICI resistance? (4) Which combination regimens can increase the share of patients benefiting from ICI? (5) Which patient/tumor characteristics predict response and its duration? Finally, (6) which timing of ICI therapy, that is, the neoadjuvant, adjuvant or therapeutic setting, offers the optimal overall outcomes for patients with MCC? A number of recent studies provide initial, but unfortunately not yet definitive answers.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145966237","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
Mutant calreticulin enables potent and selective CAR-T cell therapy in preclinical models of myeloproliferative neoplasms. 突变钙调蛋白在骨髓增生性肿瘤的临床前模型中实现了强效和选择性CAR-T细胞治疗。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-13 DOI: 10.1136/jitc-2025-012706
Cecilia Pesini, Mario Gil-Bellido, Lorena S Millan, Carmen Oñate, Adanays Calvo-Pérez, Llipsy Santiago, Eldris Iglesias, Jorge Paúl Bernal, Miguel Araujo-Voces, Laura Paz Artigas, Laura García-Martínez, Francisco J Roig, Nieves Movilla Meno, José Manuel Garcia-Aznar, Bárbara Menéndez-Jandula, María Teresa Olave, Gemma Azaceta Reinares, Marta Garrote, Alberto Alvarez-Larrán, Eva M Gálvez, Diego Sánchez Martínez, Maykel A Arias, Julian Pardo, Ariel Ramirez-Labrada

Background: The adoptive transfer of T cells engineered to express chimeric antigen receptors (CAR-T) has shown high efficacy and safety in treating various hematologic malignancies. However, many hematologic disorders, such as BCR::ABL1-negative myeloproliferative neoplasms (MPNs), lack effective treatment options. Some of these neoplasms are marked by a recurrent mutation that results in the expression of mutant calreticulin (mCALR), a neoantigen absent in healthy tissues, making it a highly specific and appealing target for CAR-T cell therapy.

Methods: Five distinct CARs were designed based on available monoclonal antibody sequences that target mCALR and were subsequently used to generate CAR-T cells. The most effective construct was selected through functional in vitro assays against mCALR-positive cell lines. Its efficacy was then evaluated in cell lines, patient-derived cells, and orthotopic xenograft models, assessing tumor burden, CAR-T cell infiltration, and animal survival. Bulk and single-cell RNA sequencing were performed on patient-derived cells and residual tumor cells from CART-treated mice, respectively, to investigate potential resistance mechanisms. The impact of the most relevant pathway alteration on CAR-T efficacy was also analyzed. Pharmacological rescue assays using targeted agents were then conducted.

Results: Among the five constructs, one demonstrated superior and specific cytotoxicity against mCALR-expressing cells, with no activity against mCALR-negative controls. This CAR-T cell also eliminated patient-derived MPN cells and controlled disease progression in xenograft models, which correlated with the persistence of CAR-T cells and tumor infiltration. Transcriptomic profiling of patient samples and residual tumor cells in spleens of treated mice revealed upregulation of anti-apoptotic proteins. Functional assays confirmed reduced CAR-T efficacy in Bcl-2 high cells, which was restored by co-treatment with venetoclax, indicating a viable combination approach to overcome resistance.

Conclusions: This study demonstrates, for the first time, the successful targeting of mCALR with CAR-T cells as a therapeutic strategy for MPNs. The chosen construct shows strong preclinical efficacy against established cell lines and patient-derived cells. Additionally, transcriptomic profiling uncovered apoptosis resistance mechanisms and supports a combination strategy with BH3 mimetics, such as venetoclax. These findings provide a compelling rationale for ongoing preclinical development and future clinical application of anti-mCALR CAR-T cells for the treatment of MPNs.

背景:嵌合抗原受体(CAR-T)修饰的T细胞过继转移在治疗多种血液系统恶性肿瘤中显示出高疗效和安全性。然而,许多血液疾病,如BCR:: abl1阴性骨髓增生性肿瘤(mpn),缺乏有效的治疗选择。这些肿瘤中的一些以复发性突变为特征,导致突变钙网蛋白(mCALR)的表达,这是一种健康组织中不存在的新抗原,使其成为CAR-T细胞治疗的高度特异性和吸引人的靶点。方法:基于现有的靶向mCALR的单克隆抗体序列设计了五种不同的CAR-T细胞,随后用于生成CAR-T细胞。通过对mcalr阳性细胞系的体外功能试验选择最有效的构建体。然后在细胞系、患者来源的细胞和原位异种移植模型中评估其疗效,评估肿瘤负荷、CAR-T细胞浸润和动物存活率。分别对患者来源的细胞和cart治疗小鼠的残余肿瘤细胞进行了大量和单细胞RNA测序,以研究潜在的耐药机制。我们还分析了最相关的通路改变对CAR-T疗效的影响。然后进行靶向药物的药理拯救试验。结果:在五个构建体中,有一个对表达mcalr的细胞表现出优越和特异性的细胞毒性,而对mcalr阴性对照没有活性。在异种移植模型中,这种CAR-T细胞也消除了患者来源的MPN细胞,并控制了疾病进展,这与CAR-T细胞的持久性和肿瘤浸润相关。患者样本和治疗小鼠脾脏残余肿瘤细胞的转录组学分析显示抗凋亡蛋白上调。功能分析证实,Bcl-2高水平细胞的CAR-T疗效降低,但与venetoclax联合治疗后,Bcl-2高水平细胞的CAR-T疗效得以恢复,这表明一种可行的联合治疗方法可以克服耐药性。结论:这项研究首次证明了CAR-T细胞靶向mCALR作为mpn的治疗策略是成功的。所选择的结构对已建立的细胞系和患者来源的细胞具有很强的临床前功效。此外,转录组学分析揭示了细胞凋亡抵抗机制,并支持与BH3模拟物(如venetoclax)的联合策略。这些发现为抗mcalr CAR-T细胞治疗mpn的临床前开发和未来临床应用提供了令人信服的理论依据。
{"title":"Mutant calreticulin enables potent and selective CAR-T cell therapy in preclinical models of myeloproliferative neoplasms.","authors":"Cecilia Pesini, Mario Gil-Bellido, Lorena S Millan, Carmen Oñate, Adanays Calvo-Pérez, Llipsy Santiago, Eldris Iglesias, Jorge Paúl Bernal, Miguel Araujo-Voces, Laura Paz Artigas, Laura García-Martínez, Francisco J Roig, Nieves Movilla Meno, José Manuel Garcia-Aznar, Bárbara Menéndez-Jandula, María Teresa Olave, Gemma Azaceta Reinares, Marta Garrote, Alberto Alvarez-Larrán, Eva M Gálvez, Diego Sánchez Martínez, Maykel A Arias, Julian Pardo, Ariel Ramirez-Labrada","doi":"10.1136/jitc-2025-012706","DOIUrl":"https://doi.org/10.1136/jitc-2025-012706","url":null,"abstract":"<p><strong>Background: </strong>The adoptive transfer of T cells engineered to express chimeric antigen receptors (CAR-T) has shown high efficacy and safety in treating various hematologic malignancies. However, many hematologic disorders, such as BCR::ABL1-negative myeloproliferative neoplasms (MPNs), lack effective treatment options. Some of these neoplasms are marked by a recurrent mutation that results in the expression of mutant calreticulin (mCALR), a neoantigen absent in healthy tissues, making it a highly specific and appealing target for CAR-T cell therapy.</p><p><strong>Methods: </strong>Five distinct CARs were designed based on available monoclonal antibody sequences that target mCALR and were subsequently used to generate CAR-T cells. The most effective construct was selected through functional in vitro assays against mCALR-positive cell lines. Its efficacy was then evaluated in cell lines, patient-derived cells, and orthotopic xenograft models, assessing tumor burden, CAR-T cell infiltration, and animal survival. Bulk and single-cell RNA sequencing were performed on patient-derived cells and residual tumor cells from CART-treated mice, respectively, to investigate potential resistance mechanisms. The impact of the most relevant pathway alteration on CAR-T efficacy was also analyzed. Pharmacological rescue assays using targeted agents were then conducted.</p><p><strong>Results: </strong>Among the five constructs, one demonstrated superior and specific cytotoxicity against mCALR-expressing cells, with no activity against mCALR-negative controls. This CAR-T cell also eliminated patient-derived MPN cells and controlled disease progression in xenograft models, which correlated with the persistence of CAR-T cells and tumor infiltration. Transcriptomic profiling of patient samples and residual tumor cells in spleens of treated mice revealed upregulation of anti-apoptotic proteins. Functional assays confirmed reduced CAR-T efficacy in Bcl-2 high cells, which was restored by co-treatment with venetoclax, indicating a viable combination approach to overcome resistance.</p><p><strong>Conclusions: </strong>This study demonstrates, for the first time, the successful targeting of mCALR with CAR-T cells as a therapeutic strategy for MPNs. The chosen construct shows strong preclinical efficacy against established cell lines and patient-derived cells. Additionally, transcriptomic profiling uncovered apoptosis resistance mechanisms and supports a combination strategy with BH3 mimetics, such as venetoclax. These findings provide a compelling rationale for ongoing preclinical development and future clinical application of anti-mCALR CAR-T cells for the treatment of MPNs.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145966259","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
Decoding adipocyte heterogeneity through single-nucleus transcriptomics unveils subtype-specific adipocytes orchestrate immunosuppressive niches in breast cancer. 通过单核转录组学解码脂肪细胞异质性揭示了乳腺癌中亚型特异性脂肪细胞协调免疫抑制壁龛。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-13 DOI: 10.1136/jitc-2025-012711
Yiwei Tong, Zheng Wang, Niu Qiao, Renhong Huang, Chenghui Wu, Haoyu Wang, Xiaochun Fei, Kunwei Shen, Xiaosong Chen

Background: While adipose tissue constitutes a substantial proportion of breast composition, the functional characteristics and pathological relevance of the adipocyte microenvironment in breast carcinogenesis remain undercharacterized. This study employs single-nucleus RNA sequencing (snRNA-seq) to establish a comprehensive cellular atlas of adipocyte heterogeneity across molecular subtypes of breast cancer, aiming to elucidate subtype-specific adipocyte contributions to tumor microenvironment modulation.

Methods: snRNA-seq was performed on breast adipose tissues isolated from individuals without cancer and treatment-naïve breast cancer. Various adipocyte and pre-adipocyte subclusters were identified. Comparative analyses of cellular distribution and transcriptional profiles were performed across disease states and molecular subtypes. Pseudotime, cell communication, and immunofluorescence analyses were further implemented to investigate cellular dynamics and microenvironment interactions.

Results: snRNA-seq data of 86,529 nuclei were obtained. Three adipocyte and seven pre-adipocyte subclusters were identified, of which Adi_LDLR, Pre_Adi_LDLR, and Pre_Adi_LGR4_TGFBR1 were restricted to cancer-associated adipose (CAAs). Adi_LDLR and Pre_Adi_LDLR were enriched in estrogen receptor (ER)-positive CAAs and related to cell senescence and immunosuppression. Pre_Adi_LGR4_TGFBR1 was predominantly present in triple-negative breast cancer, functionally pro-proliferative, immunosuppressive, and lacked normal adipose function. The immunofluorescence intensity of LDLR (p=0.031) and TGFBR1 (p=0.038) was positively associated with disease recurrence, suggesting the formation of immunosuppressive niches by these cancer-specific adipose subsets in both subtypes. Cell communication analyses revealed a specific (pre-) adipocyte-macrophage interaction via ligand-receptor pairs involved in stromal remodeling and tumor migration for ER-positive tumors, whereas tumor proliferation and metastasis for triple-negative ones likely contribute to tumor progression.

Conclusions: This study delineated a distinct adipocyte landscape in breast cancer and subtype-specific immunosuppressive niches fostered by CAAs and (pre-) adipocyte-macrophage interactions. These findings provide novel therapeutic targets for microenvironment-directed interventions in breast oncology.

背景:虽然脂肪组织在乳房组成中占很大比例,但脂肪细胞微环境在乳腺癌发生中的功能特征和病理相关性仍然不清楚。本研究采用单核RNA测序(snRNA-seq)建立了乳腺癌分子亚型中脂肪细胞异质性的综合细胞图谱,旨在阐明亚型特异性脂肪细胞对肿瘤微环境调节的贡献。方法:对无癌个体和treatment-naïve乳腺癌个体分离的乳腺脂肪组织进行snRNA-seq检测。鉴定出各种脂肪细胞和前脂肪细胞亚群。对不同疾病状态和分子亚型的细胞分布和转录谱进行了比较分析。伪时间、细胞通讯和免疫荧光分析进一步用于研究细胞动力学和微环境相互作用。结果:共获得86529个snRNA-seq数据。鉴定出3个脂肪细胞和7个前脂肪细胞亚簇,其中Adi_LDLR、Pre_Adi_LDLR和Pre_Adi_LGR4_TGFBR1仅限于癌症相关脂肪(CAAs)。Adi_LDLR和Pre_Adi_LDLR在雌激素受体(ER)阳性CAAs中富集,与细胞衰老和免疫抑制有关。Pre_Adi_LGR4_TGFBR1主要存在于三阴性乳腺癌中,功能上促增殖,免疫抑制,缺乏正常的脂肪功能。LDLR (p=0.031)和TGFBR1 (p=0.038)的免疫荧光强度与疾病复发呈正相关,提示两种亚型中这些癌症特异性脂肪亚群形成了免疫抑制生态位。细胞通讯分析显示,在er阳性肿瘤中,脂肪细胞-巨噬细胞通过配体-受体对进行特异性(前)相互作用,参与基质重塑和肿瘤迁移,而三阴性肿瘤的肿瘤增殖和转移可能有助于肿瘤进展。结论:这项研究描绘了乳腺癌中独特的脂肪细胞景观和CAAs和(前)脂肪细胞-巨噬细胞相互作用培养的亚型特异性免疫抑制生态位。这些发现为乳腺肿瘤微环境干预提供了新的治疗靶点。
{"title":"Decoding adipocyte heterogeneity through single-nucleus transcriptomics unveils subtype-specific adipocytes orchestrate immunosuppressive niches in breast cancer.","authors":"Yiwei Tong, Zheng Wang, Niu Qiao, Renhong Huang, Chenghui Wu, Haoyu Wang, Xiaochun Fei, Kunwei Shen, Xiaosong Chen","doi":"10.1136/jitc-2025-012711","DOIUrl":"https://doi.org/10.1136/jitc-2025-012711","url":null,"abstract":"<p><strong>Background: </strong>While adipose tissue constitutes a substantial proportion of breast composition, the functional characteristics and pathological relevance of the adipocyte microenvironment in breast carcinogenesis remain undercharacterized. This study employs single-nucleus RNA sequencing (snRNA-seq) to establish a comprehensive cellular atlas of adipocyte heterogeneity across molecular subtypes of breast cancer, aiming to elucidate subtype-specific adipocyte contributions to tumor microenvironment modulation.</p><p><strong>Methods: </strong>snRNA-seq was performed on breast adipose tissues isolated from individuals without cancer and treatment-naïve breast cancer. Various adipocyte and pre-adipocyte subclusters were identified. Comparative analyses of cellular distribution and transcriptional profiles were performed across disease states and molecular subtypes. Pseudotime, cell communication, and immunofluorescence analyses were further implemented to investigate cellular dynamics and microenvironment interactions.</p><p><strong>Results: </strong>snRNA-seq data of 86,529 nuclei were obtained. Three adipocyte and seven pre-adipocyte subclusters were identified, of which Adi_LDLR, Pre_Adi_LDLR, and Pre_Adi_LGR4_TGFBR1 were restricted to cancer-associated adipose (CAAs). Adi_LDLR and Pre_Adi_LDLR were enriched in estrogen receptor (ER)-positive CAAs and related to cell senescence and immunosuppression. Pre_Adi_LGR4_TGFBR1 was predominantly present in triple-negative breast cancer, functionally pro-proliferative, immunosuppressive, and lacked normal adipose function. The immunofluorescence intensity of LDLR (p=0.031) and TGFBR1 (p=0.038) was positively associated with disease recurrence, suggesting the formation of immunosuppressive niches by these cancer-specific adipose subsets in both subtypes. Cell communication analyses revealed a specific (pre-) adipocyte-macrophage interaction via ligand-receptor pairs involved in stromal remodeling and tumor migration for ER-positive tumors, whereas tumor proliferation and metastasis for triple-negative ones likely contribute to tumor progression.</p><p><strong>Conclusions: </strong>This study delineated a distinct adipocyte landscape in breast cancer and subtype-specific immunosuppressive niches fostered by CAAs and (pre-) adipocyte-macrophage interactions. These findings provide novel therapeutic targets for microenvironment-directed interventions in breast oncology.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145966274","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
Cell therapy in sarcoma: current landscape and future directions. 肉瘤的细胞治疗:现状和未来方向。
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-12 DOI: 10.1136/jitc-2025-013396
Taha Koray Sahin, Theodora Germetaki, Deniz Can Guven, Serkan Akin, Omer Dizdar, Fiona Thistlethwaite, Elizabeth A Connolly, Kok Haw Jonathan Lim

Sarcomas are rare malignancies of mesenchymal origin, characterized by significant biological and clinical heterogeneity. Many subtypes demonstrate limited sensitivity to standard systemic treatments, including immune checkpoint inhibitors. Cell therapy has emerged as a promising strategy, with the potential of durable clinical responses seen with genetically-engineered T-cell receptor T-cell therapies (TCR-T) such as those targeting the cancer-testis antigen MAGE-A4 in synovial sarcoma, leading to the US Food and Drug Administration approval of afamitresgene autoleucel in 2024. This constituted only the second approval of a cell therapy in a solid tumor following lifileucel in melanoma and demonstrated the potential of cell therapies in sarcomas. This review provides the current landscape and growing potential of cell therapies in sarcomas, including TCR-T, chimeric antigen receptor-T cells, tumor-infiltrating lymphocytes, natural killer (NK) cells, and mesenchymal stromal cells. However, the broader application of these therapies is hindered by the lack of targetable sarcoma-restricted immunogenic epitopes, spatiotemporal intratumoral heterogeneity, and a profoundly immunosuppressive tumor microenvironment that impedes effector-cell trafficking, expansion and persistence. While cell therapies hold promise for integration into precision medicine approaches for sarcomas, their successful implementation will require careful evaluation of clinical feasibility, logistical considerations and cost-effectiveness to optimize patient outcomes.

肉瘤是一种罕见的间充质恶性肿瘤,具有显著的生物学和临床异质性。许多亚型对包括免疫检查点抑制剂在内的标准全身治疗表现出有限的敏感性。细胞疗法已经成为一种很有前途的策略,具有持久临床反应的潜力的基因工程t细胞受体t细胞疗法(TCR-T),如针对滑膜肉瘤的癌睾丸抗原MAGE-A4的疗法,导致美国食品和药物管理局于2024年批准了afamitresgene autoeucel。这是继lifileucel治疗黑色素瘤之后,第二次批准细胞疗法治疗实体瘤,并证明了细胞疗法治疗肉瘤的潜力。本文综述了肿瘤细胞治疗的现状和发展潜力,包括TCR-T、嵌合抗原受体- t细胞、肿瘤浸润淋巴细胞、自然杀伤(NK)细胞和间充质基质细胞。然而,这些疗法的广泛应用受到缺乏靶向性肉瘤限制免疫原性表位、肿瘤内时空异质性和严重的免疫抑制肿瘤微环境的阻碍,这些微环境阻碍了效应细胞的运输、扩展和持续。虽然细胞疗法有望整合到肉瘤的精准医学方法中,但它们的成功实施需要仔细评估临床可行性、后勤考虑和成本效益,以优化患者的治疗结果。
{"title":"Cell therapy in sarcoma: current landscape and future directions.","authors":"Taha Koray Sahin, Theodora Germetaki, Deniz Can Guven, Serkan Akin, Omer Dizdar, Fiona Thistlethwaite, Elizabeth A Connolly, Kok Haw Jonathan Lim","doi":"10.1136/jitc-2025-013396","DOIUrl":"https://doi.org/10.1136/jitc-2025-013396","url":null,"abstract":"<p><p>Sarcomas are rare malignancies of mesenchymal origin, characterized by significant biological and clinical heterogeneity. Many subtypes demonstrate limited sensitivity to standard systemic treatments, including immune checkpoint inhibitors. Cell therapy has emerged as a promising strategy, with the potential of durable clinical responses seen with genetically-engineered T-cell receptor T-cell therapies (TCR-T) such as those targeting the cancer-testis antigen MAGE-A4 in synovial sarcoma, leading to the US Food and Drug Administration approval of afamitresgene autoleucel in 2024. This constituted only the second approval of a cell therapy in a solid tumor following lifileucel in melanoma and demonstrated the potential of cell therapies in sarcomas. This review provides the current landscape and growing potential of cell therapies in sarcomas, including TCR-T, chimeric antigen receptor-T cells, tumor-infiltrating lymphocytes, natural killer (NK) cells, and mesenchymal stromal cells. However, the broader application of these therapies is hindered by the lack of targetable sarcoma-restricted immunogenic epitopes, spatiotemporal intratumoral heterogeneity, and a profoundly immunosuppressive tumor microenvironment that impedes effector-cell trafficking, expansion and persistence. While cell therapies hold promise for integration into precision medicine approaches for sarcomas, their successful implementation will require careful evaluation of clinical feasibility, logistical considerations and cost-effectiveness to optimize patient outcomes.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145959450","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
Serum cytokines predict response and survival in esophageal squamous cell carcinoma receiving chemoradiotherapy combined with anti-PD-1 antibody: analyses of two phase II clinical trials. 血清细胞因子预测食管鳞状细胞癌接受放化疗联合抗pd -1抗体的反应和生存:两项II期临床试验的分析
IF 10.6 1区 医学 Q1 IMMUNOLOGY Pub Date : 2026-01-12 DOI: 10.1136/jitc-2025-013065
BaoQing Chen, Junying Chen, Sifen Wang, Kunhao Bai, Zimeng Li, Biqi Chen, Ruixi Wang, Xingyuan Cheng, Yilu Gao, Chen Yi, Peiying Cen, Shuangjiang Li, Mihnea P Dragomir, Yujia Zhu, Qiaoqiao Li, Hong Yang, Mian Xi

Purpose: Chemoradiotherapy (CRT) combined with anti-PD-1 for locally advanced esophageal squamous cell carcinoma (ESCC) has shown promising efficacy but lack the predictive biomarkers to identify patients who could benefit from this therapy. The predictive value of serum cytokines in ESCC patients remains unclear. We aimed to identify cytokine-based biomarkers for treatment response and survival in this setting.

Experimental design: Exploratory analyses were conducted on 81 ESCC patients from two phase II trials treated with CRT plus toripalimab, with validation in an independent prospective cohort (n=61). Nineteen serum cytokines were assessed at baseline, during, and post-CRT plus anti-PD-1 antibody. A cytokine-based risk score model (CYTOscore) was constructed. Multi-omics profiling including RNA-seq, WES, and spatial transcriptomics were performed to explore potential differences in tumor microenvironments.

Results: Cox analyses identified Interleukin-8 (IL-8), C-C motif chemokine ligand 3 (CCL3), and C-C motif chemokine ligand 4 (CCL4) as potential biomarkers and were used to constructed the CYTOscore. Patients stratified by baseline CYTOscore showed significantly longer OS (HR, 0.31; 95%CI, 0.16-0.62; p= 0.00045) and PFS (HR, 0.33; 95%CI, 0.17-0.62; p= 0.00036) in the low-risk group, which also had higher complete response (CR) rates (66% vs 35%, p=0.014). These finding were next validated in the external cohort, with the low-risk group demonstrating higher CR rates (66% vs 27%, p=0.039) and longer OS (HR 0.30, 95% CI 0.09-0.99, p=0.045). A nomogram incorporating baseline CYTOscore and clinical characteristics showed promising predictive accuracy in 1-, 2-, and 3-year OS (AUC=0.77, 0.78, and 0.76). Multi-omics analysis revealed enriched interferon-γ/α signaling in B cells within low-risk patients.

Conclusions: The CYTOscore based on IL-8, CCL3, and CCL4 effectively predicts treatment response and survival in ESCC patients receiving CRT plus anti-PD-1 antibody.

目的:放化疗(CRT)联合抗pd -1治疗局部晚期食管鳞状细胞癌(ESCC)显示出有希望的疗效,但缺乏预测性生物标志物来识别可以从这种治疗中获益的患者。血清细胞因子在ESCC患者中的预测价值尚不清楚。我们的目的是确定基于细胞因子的生物标志物在这种情况下的治疗反应和生存。实验设计:对来自两项II期试验的81例ESCC患者进行了探索性分析,并在独立的前瞻性队列(n=61)中进行了验证。19种血清细胞因子在基线、crt期间和crt后加上抗pd -1抗体进行评估。构建基于细胞因子的风险评分模型(CYTOscore)。多组学分析包括RNA-seq、WES和空间转录组学来探索肿瘤微环境的潜在差异。结果:Cox分析发现白细胞介素-8 (IL-8)、C-C基序趋化因子配体3 (CCL3)和C-C基序趋化因子配体4 (CCL4)是潜在的生物标志物,并用于构建细胞评分。根据基线CYTOscore分层的患者显示,低危组的OS (HR, 0.31; 95%CI, 0.16-0.62; p= 0.00045)和PFS (HR, 0.33; 95%CI, 0.17-0.62; p= 0.00036)明显更长,完全缓解率(CR)也更高(66% vs 35%, p=0.014)。这些发现随后在外部队列中得到验证,低风险组表现出更高的CR率(66%对27%,p=0.039)和更长的OS (HR 0.30, 95% CI 0.09-0.99, p=0.045)。结合基线细胞评分和临床特征的nomogram预测1年、2年和3年OS的准确性(AUC=0.77、0.78和0.76)。多组学分析显示低危患者B细胞中干扰素γ/α信号富集。结论:基于IL-8、CCL3和CCL4的细胞评分可有效预测接受CRT +抗pd -1抗体治疗的ESCC患者的治疗反应和生存期。
{"title":"Serum cytokines predict response and survival in esophageal squamous cell carcinoma receiving chemoradiotherapy combined with anti-PD-1 antibody: analyses of two phase II clinical trials.","authors":"BaoQing Chen, Junying Chen, Sifen Wang, Kunhao Bai, Zimeng Li, Biqi Chen, Ruixi Wang, Xingyuan Cheng, Yilu Gao, Chen Yi, Peiying Cen, Shuangjiang Li, Mihnea P Dragomir, Yujia Zhu, Qiaoqiao Li, Hong Yang, Mian Xi","doi":"10.1136/jitc-2025-013065","DOIUrl":"https://doi.org/10.1136/jitc-2025-013065","url":null,"abstract":"<p><strong>Purpose: </strong>Chemoradiotherapy (CRT) combined with anti-PD-1 for locally advanced esophageal squamous cell carcinoma (ESCC) has shown promising efficacy but lack the predictive biomarkers to identify patients who could benefit from this therapy. The predictive value of serum cytokines in ESCC patients remains unclear. We aimed to identify cytokine-based biomarkers for treatment response and survival in this setting.</p><p><strong>Experimental design: </strong>Exploratory analyses were conducted on 81 ESCC patients from two phase II trials treated with CRT plus toripalimab, with validation in an independent prospective cohort (n=61). Nineteen serum cytokines were assessed at baseline, during, and post-CRT plus anti-PD-1 antibody. A cytokine-based risk score model (CYTOscore) was constructed. Multi-omics profiling including RNA-seq, WES, and spatial transcriptomics were performed to explore potential differences in tumor microenvironments.</p><p><strong>Results: </strong>Cox analyses identified Interleukin-8 (IL-8), C-C motif chemokine ligand 3 (CCL3), and C-C motif chemokine ligand 4 (CCL4) as potential biomarkers and were used to constructed the CYTOscore. Patients stratified by baseline CYTOscore showed significantly longer OS (HR, 0.31; 95%CI, 0.16-0.62; p= 0.00045) and PFS (HR, 0.33; 95%CI, 0.17-0.62; p= 0.00036) in the low-risk group, which also had higher complete response (CR) rates (66% vs 35%, p=0.014). These finding were next validated in the external cohort, with the low-risk group demonstrating higher CR rates (66% vs 27%, p=0.039) and longer OS (HR 0.30, 95% CI 0.09-0.99, p=0.045). A nomogram incorporating baseline CYTOscore and clinical characteristics showed promising predictive accuracy in 1-, 2-, and 3-year OS (AUC=0.77, 0.78, and 0.76). Multi-omics analysis revealed enriched interferon-γ/α signaling in B cells within low-risk patients.</p><p><strong>Conclusions: </strong>The CYTOscore based on IL-8, CCL3, and CCL4 effectively predicts treatment response and survival in ESCC patients receiving CRT plus anti-PD-1 antibody.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"14 1","pages":""},"PeriodicalIF":10.6,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145959412","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
Journal for Immunotherapy of Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1