首页 > 最新文献

Journal for Immunotherapy of Cancer最新文献

英文 中文
Spatial and single-cell transcriptomics reveal cellular heterogeneity and a novel cancer-promoting Treg cell subset in human clear-cell renal cell carcinoma. 空间和单细胞转录组学揭示了人类透明细胞肾细胞癌的细胞异质性和一种新的促癌Treg细胞亚群。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-04 DOI: 10.1136/jitc-2024-010183
Xiyu Song, Yumeng Zhu, Wenwen Geng, Jianhua Jiao, Hongjiao Liu, Ruo Chen, Qian He, Lijuan Wang, Xiuxuan Sun, Weijun Qin, Jiejie Geng, Zhinan Chen

Background: Clear cell renal cell carcinoma (ccRCC) is the most common histologic type of RCC. However, the spatial and functional heterogeneity of immunosuppressive cells and the mechanisms by which their interactions promote immunosuppression in the ccRCC have not been thoroughly investigated.

Methods: To further investigate the cellular and regional heterogeneity of ccRCC, we analyzed single-cell and spatial transcriptome RNA sequencing data from four patients, which were obtained from samples from multiple regions, including the tumor core, tumor-normal interface, and distal normal tissue. On the basis, the findings were investigated in vitro using tissue and blood samples from 15 patients with ccRCC and validated in the broader samples on tissue microarrays.

Results: In this study, we revealed previously unreported subsets of both stromal and immune cells, as well as mapped their spatial location at finer resolution. In addition, we validated the clusters of tumor cells after removing batch effects according to six characterized gene sets, including epithelial-mesenchymal transitionhigh clusters, metastatic clusters and proximal tubulehigh clusters. Importantly, we identified a special regulatory T (Treg) cell subpopulation that has the molecular characteristics of terminal effector Treg cells but expresses multiple cytokines, such as interleukin (IL)-1β and IL-18. This group of Treg cells has stronger immunosuppressive function and was associated with a worse prognosis in ccRCC cohorts. They were colocalized with MRC1 + FOLR2 + tumor-associated macrophages (TAMs) at the tumor-normal interface to form a positive feedback loop, maintaining a synergistic procarcinogenic effect. In addition, we traced the origin of IL-1β+ Treg cells and revealed that IL-18 can induce the expression of IL-1β in Treg cells via the ERK/NF-κB pathway.

Conclusions: We demonstrated a novel cancer-promoting Treg cell subset and its interactions with MRC1 + FOLR2 +TAMs, which provides new insight into Treg cell heterogeneity and potential therapeutic targets for ccRCC.

背景:透明细胞肾细胞癌(ccRCC)是肾癌最常见的组织学类型。然而,免疫抑制细胞的空间和功能异质性及其相互作用促进ccRCC免疫抑制的机制尚未得到深入研究。方法:为了进一步研究ccRCC的细胞和区域异质性,我们分析了4例患者的单细胞和空间转录组RNA测序数据,这些数据来自肿瘤核心、肿瘤-正常界面和远端正常组织等多个区域的样本。在此基础上,研究人员利用15例ccRCC患者的组织和血液样本在体外进行了研究,并在组织微阵列上对更广泛的样本进行了验证。结果:在这项研究中,我们揭示了以前未报道的基质细胞和免疫细胞亚群,并以更精细的分辨率绘制了它们的空间位置。此外,我们根据六个特征基因集验证了去除批效应后的肿瘤细胞簇,包括上皮-间充质过渡高簇、转移性簇和近端小管高簇。重要的是,我们发现了一个特殊的调节性T (Treg)细胞亚群,它具有末端效应Treg细胞的分子特征,但表达多种细胞因子,如白细胞介素(IL)-1β和IL-18。这组Treg细胞具有较强的免疫抑制功能,在ccRCC队列中与较差的预后相关。它们与MRC1 + FOLR2 +肿瘤相关巨噬细胞(tam)在肿瘤-正常界面共定位,形成正反馈回路,保持协同的前致癌作用。此外,我们追踪了IL-1β+ Treg细胞的来源,发现IL-18可以通过ERK/NF-κB途径诱导IL-1β在Treg细胞中的表达。结论:我们展示了一种新的促癌Treg细胞亚群及其与MRC1 + FOLR2 + tam的相互作用,这为Treg细胞异质性和ccRCC的潜在治疗靶点提供了新的见解。
{"title":"Spatial and single-cell transcriptomics reveal cellular heterogeneity and a novel cancer-promoting Treg cell subset in human clear-cell renal cell carcinoma.","authors":"Xiyu Song, Yumeng Zhu, Wenwen Geng, Jianhua Jiao, Hongjiao Liu, Ruo Chen, Qian He, Lijuan Wang, Xiuxuan Sun, Weijun Qin, Jiejie Geng, Zhinan Chen","doi":"10.1136/jitc-2024-010183","DOIUrl":"10.1136/jitc-2024-010183","url":null,"abstract":"<p><strong>Background: </strong>Clear cell renal cell carcinoma (ccRCC) is the most common histologic type of RCC. However, the spatial and functional heterogeneity of immunosuppressive cells and the mechanisms by which their interactions promote immunosuppression in the ccRCC have not been thoroughly investigated.</p><p><strong>Methods: </strong>To further investigate the cellular and regional heterogeneity of ccRCC, we analyzed single-cell and spatial transcriptome RNA sequencing data from four patients, which were obtained from samples from multiple regions, including the tumor core, tumor-normal interface, and distal normal tissue. On the basis, the findings were investigated in vitro using tissue and blood samples from 15 patients with ccRCC and validated in the broader samples on tissue microarrays.</p><p><strong>Results: </strong>In this study, we revealed previously unreported subsets of both stromal and immune cells, as well as mapped their spatial location at finer resolution. In addition, we validated the clusters of tumor cells after removing batch effects according to six characterized gene sets, including epithelial-mesenchymal transition<sup>high</sup> clusters, metastatic clusters and proximal tubule<sup>high</sup> clusters. Importantly, we identified a special regulatory T (Treg) cell subpopulation that has the molecular characteristics of terminal effector Treg cells but expresses multiple cytokines, such as interleukin (IL)-1β and IL-18. This group of Treg cells has stronger immunosuppressive function and was associated with a worse prognosis in ccRCC cohorts. They were colocalized with <i>MRC1</i> <sup>+</sup> <i>FOLR2</i> <sup>+</sup> tumor-associated macrophages (TAMs) at the tumor-normal interface to form a positive feedback loop, maintaining a synergistic procarcinogenic effect. In addition, we traced the origin of IL-1β<sup>+</sup> Treg cells and revealed that IL-18 can induce the expression of IL-1β in Treg cells via the ERK/NF-κB pathway.</p><p><strong>Conclusions: </strong>We demonstrated a novel cancer-promoting Treg cell subset and its interactions with <i>MRC1</i> <sup>+</sup> <i>FOLR2</i> <sup>+</sup>TAMs, which provides new insight into Treg cell heterogeneity and potential therapeutic targets for ccRCC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3,"publicationDate":"2025-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11748785/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927112","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
Correction: Associations of concomitant medications with immune-related adverse events and survival in advanced cancers treated with immune checkpoint inhibitors: a comprehensive pan-cancer analysis. 修正:在免疫检查点抑制剂治疗的晚期癌症中,伴随药物与免疫相关不良事件和生存率的关联:一项全面的泛癌症分析。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-31 DOI: 10.1136/jitc-2024-008806corr1
{"title":"Correction: Associations of concomitant medications with immune-related adverse events and survival in advanced cancers treated with immune checkpoint inhibitors: a comprehensive pan-cancer analysis.","authors":"","doi":"10.1136/jitc-2024-008806corr1","DOIUrl":"https://doi.org/10.1136/jitc-2024-008806corr1","url":null,"abstract":"","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142909544","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 atezolizumab + chemotherapy for resectable NSCLC: 3-year clinical update of phase II clinical trial results and translational findings. 可切除NSCLC的新辅助atezolizumab +化疗:3年II期临床试验结果和转化结果的临床更新
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-25 DOI: 10.1136/jitc-2024-009301
Brian S Henick, Peter D Koch, Justin F Gainor, Mark M Awad, Codruta Chiuzan, Stephanie Izard, Yohanna Georgis, Samyukta Mallick, Robert F Garofano, Cheryl V Wong, Anjali Saqi, Jessica Grindheim, Katja Schulze, Joshua R Sonett, Naiyer A Rizvi, Benjamin Izar, Alison M Taylor, Catherine A Shu

Introduction: Neoadjuvant chemoimmunotherapy has achieved overall survival (OS) benefit for patients with resectable non-small cell lung cancer (NSCLC). Here, we present outcomes after 3 years of follow-up from the first reported study of neoadjuvant atezolizumab+chemotherapy.

Methods: This open-label, multicenter single-arm investigator-initiated phase II study conducted at three US hospitals tested up to four cycles of atezolizumab, carboplatin, and nab-paclitaxel prior to surgery. Major pathological response (MPR, primary endpoint) was previously reported; here, we report 3-year disease-free survival (DFS), OS, and clinical characteristics of patients developing brain metastases (BM) with integrated data from tumor genomics, gene expression, and quantitative immunofluorescent measurement of immune markers.

Results: Of 30 enrolled patients, 29 were taken to the operating room. 26 underwent R0 resection, with 17 experiencing MPR (10 pCR). With a median follow-up of 39.5 months, the median OS was 55.8 months, and the median DFS was 34.5 months. Landmark OS at 36 months was 77%. Among 14 patients with recurrent disease, 6 patients had BM. Patients whose tumors had mutations in STK11 and KEAP1 did not have a significantly higher incidence of BM. Reduced copy number of STK11 and KEAP1, both residing on chromosome 19p, was observed in ~1/3 of tumors. Reduced CN of STK11 was significantly associated with worse pathological response and incidence of BM.

Conclusions: Consistent with recent phase III studies, 3-year OS data with neoadjuvant atezolizumab+chemotherapy was associated with prolonged PFS and OS. Establishing associations between STK11 and KEAP1 genomic alterations and key clinical outcomes in early-stage NSCLC requires further study.

新辅助化疗免疫治疗对可切除的非小细胞肺癌(NSCLC)患者的总生存期(OS)有所改善。在这里,我们介绍了首次报道的新辅助atezolizumab+化疗研究3年随访后的结果。方法:这项开放标签、多中心、单臂研究者发起的II期研究在美国三家医院进行,测试了手术前atezolizumab、卡铂和nab-紫杉醇的四个周期。主要病理反应(MPR,主要终点)先前有报道;在这里,我们通过肿瘤基因组学、基因表达和免疫标记物定量免疫荧光测量的综合数据,报告了发生脑转移(BM)患者的3年无病生存(DFS)、OS和临床特征。结果:30例入组患者中,29例进入手术室。26例R0切除,17例MPR(10例pCR)。中位随访时间为39.5个月,中位OS为55.8个月,中位DFS为34.5个月。Landmark OS在36个月时是77%。14例复发患者中有6例发生BM。肿瘤中STK11和KEAP1基因突变的患者,BM的发病率并没有显著增高。位于19p染色体上的STK11和KEAP1拷贝数减少,在约1/3的肿瘤中发现。STK11的CN降低与较差的病理反应和BM的发生率显著相关。结论:与最近的III期研究一致,新辅助atezolizumab+化疗的3年OS数据与延长的PFS和OS相关。确定STK11和KEAP1基因组改变与早期NSCLC关键临床结局之间的关系需要进一步研究。
{"title":"Neoadjuvant atezolizumab + chemotherapy for resectable NSCLC: 3-year clinical update of phase II clinical trial results and translational findings.","authors":"Brian S Henick, Peter D Koch, Justin F Gainor, Mark M Awad, Codruta Chiuzan, Stephanie Izard, Yohanna Georgis, Samyukta Mallick, Robert F Garofano, Cheryl V Wong, Anjali Saqi, Jessica Grindheim, Katja Schulze, Joshua R Sonett, Naiyer A Rizvi, Benjamin Izar, Alison M Taylor, Catherine A Shu","doi":"10.1136/jitc-2024-009301","DOIUrl":"10.1136/jitc-2024-009301","url":null,"abstract":"<p><strong>Introduction: </strong>Neoadjuvant chemoimmunotherapy has achieved overall survival (OS) benefit for patients with resectable non-small cell lung cancer (NSCLC). Here, we present outcomes after 3 years of follow-up from the first reported study of neoadjuvant atezolizumab+chemotherapy.</p><p><strong>Methods: </strong>This open-label, multicenter single-arm investigator-initiated phase II study conducted at three US hospitals tested up to four cycles of atezolizumab, carboplatin, and nab-paclitaxel prior to surgery. Major pathological response (MPR, primary endpoint) was previously reported; here, we report 3-year disease-free survival (DFS), OS, and clinical characteristics of patients developing brain metastases (BM) with integrated data from tumor genomics, gene expression, and quantitative immunofluorescent measurement of immune markers.</p><p><strong>Results: </strong>Of 30 enrolled patients, 29 were taken to the operating room. 26 underwent R0 resection, with 17 experiencing MPR (10 pCR). With a median follow-up of 39.5 months, the median OS was 55.8 months, and the median DFS was 34.5 months. Landmark OS at 36 months was 77%. Among 14 patients with recurrent disease, 6 patients had BM. Patients whose tumors had mutations in <i>STK11</i> and <i>KEAP1</i> did not have a significantly higher incidence of BM. Reduced copy number of <i>STK11</i> and <i>KEAP1</i>, both residing on chromosome 19p, was observed in ~1/3 of tumors. Reduced CN of <i>STK11</i> was significantly associated with worse pathological response and incidence of BM.</p><p><strong>Conclusions: </strong>Consistent with recent phase III studies, 3-year OS data with neoadjuvant atezolizumab+chemotherapy was associated with prolonged PFS and OS. Establishing associations between <i>STK11</i> and <i>KEAP1</i> genomic alterations and key clinical outcomes in early-stage NSCLC requires further study.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11752048/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142894412","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
Lactylation-driven TNFR2 expression in regulatory T cells promotes the progression of malignant pleural effusion. 调节性T细胞中乳酸化驱动的TNFR2表达促进恶性胸腔积液的进展。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-25 DOI: 10.1136/jitc-2024-010040
Qianqian Xue, Wenbei Peng, Siyu Zhang, Xiaoshan Wei, Linlin Ye, Zihao Wang, Xuan Xiang, Yao Liu, Haolei Wang, Qiong Zhou

Background: Although tumor necrosis factor receptor 2 (TNFR2) has been recognized as an attractive next-generation candidate target for cancer immunotherapy, the factors that regulate the gene expression and their mechanistic effects on tumor-infiltrating regulatory T cells (Treg cells) remain poorly understood.

Methods: Single-cell RNA sequencing analysis was employed to analyze the phenotypic and functional differences between TNFR2+ Treg cells and TNFR2- Treg cells. Malignant pleural effusion (MPE) from humans and mouse was used to investigate the potential mechanisms by which lactate regulates TNFR2 expression.

Results: Treg cells with high TNFR2 expression exhibited elevated levels of immune checkpoint molecules. Additionally, the high expression of TNFR2 on Treg cells was positively correlated with a poor prognosis in MPE patients. Moreover, we revealed that lactate upregulated TNFR2 expression on Treg cells, thereby enhancing their immunosuppressive function in MPE. Mechanistically, lactate modulated the gene transcription of transcription factor nuclear factor-κB p65 (NF-κB p65) through histone H3K18 lactylation (H3K18la), subsequently upregulating the gene expression of TNFR2 and expediting the progression of MPE. Notably, lactate metabolism blockade combined with immune checkpoint blockade (ICB) therapy effectively enhanced the efficacy of ICB therapy, prolonged the survival time of MPE mice, and improved immunosuppression in the microenvironment of MPE.

Conclusions: The study explains the mechanism that regulates TNFR2 expression on Treg cells and its function in MPE progression, providing novel insights into the epigenetic regulation of tumor development and metabolic strategies for MPE treatment by targeting lactate metabolism in Treg cells.

背景:尽管肿瘤坏死因子受体2 (TNFR2)已被认为是有吸引力的下一代癌症免疫治疗候选靶点,但调控基因表达的因素及其对肿瘤浸润调节性T细胞(Treg细胞)的机制作用仍然知之甚少。方法:采用单细胞RNA测序分析TNFR2+ Treg细胞和TNFR2- Treg细胞的表型和功能差异。利用人类和小鼠的恶性胸腔积液(MPE)来研究乳酸调节TNFR2表达的潜在机制。结果:高TNFR2表达的Treg细胞免疫检查点分子水平升高。此外,Treg细胞上TNFR2的高表达与MPE患者的不良预后呈正相关。此外,我们发现乳酸上调Treg细胞上TNFR2的表达,从而增强其在MPE中的免疫抑制功能。机制上,乳酸通过组蛋白H3K18乳酸化(H3K18la)调节转录因子核因子-κB p65 (NF-κB p65)的基因转录,进而上调TNFR2的基因表达,加速MPE的进展。值得注意的是,乳酸代谢阻断联合免疫检查点阻断(immune checkpoint blockade, ICB)治疗有效增强了ICB治疗的疗效,延长了MPE小鼠的生存时间,改善了MPE微环境中的免疫抑制。结论:本研究解释了Treg细胞TNFR2表达调控机制及其在MPE进展中的作用,为肿瘤发生的表观遗传调控和靶向Treg细胞乳酸代谢治疗MPE的代谢策略提供了新的见解。
{"title":"Lactylation-driven TNFR2 expression in regulatory T cells promotes the progression of malignant pleural effusion.","authors":"Qianqian Xue, Wenbei Peng, Siyu Zhang, Xiaoshan Wei, Linlin Ye, Zihao Wang, Xuan Xiang, Yao Liu, Haolei Wang, Qiong Zhou","doi":"10.1136/jitc-2024-010040","DOIUrl":"10.1136/jitc-2024-010040","url":null,"abstract":"<p><strong>Background: </strong>Although tumor necrosis factor receptor 2 (TNFR2) has been recognized as an attractive next-generation candidate target for cancer immunotherapy, the factors that regulate the gene expression and their mechanistic effects on tumor-infiltrating regulatory T cells (Treg cells) remain poorly understood.</p><p><strong>Methods: </strong>Single-cell RNA sequencing analysis was employed to analyze the phenotypic and functional differences between TNFR2<sup>+</sup> Treg cells and TNFR2<sup>-</sup> Treg cells. Malignant pleural effusion (MPE) from humans and mouse was used to investigate the potential mechanisms by which lactate regulates TNFR2 expression.</p><p><strong>Results: </strong>Treg cells with high TNFR2 expression exhibited elevated levels of immune checkpoint molecules. Additionally, the high expression of TNFR2 on Treg cells was positively correlated with a poor prognosis in MPE patients. Moreover, we revealed that lactate upregulated TNFR2 expression on Treg cells, thereby enhancing their immunosuppressive function in MPE. Mechanistically, lactate modulated the gene transcription of transcription factor nuclear factor-κB p65 (NF-κB p65) through histone H3K18 lactylation (H3K18la), subsequently upregulating the gene expression of TNFR2 and expediting the progression of MPE. Notably, lactate metabolism blockade combined with immune checkpoint blockade (ICB) therapy effectively enhanced the efficacy of ICB therapy, prolonged the survival time of MPE mice, and improved immunosuppression in the microenvironment of MPE.</p><p><strong>Conclusions: </strong>The study explains the mechanism that regulates TNFR2 expression on Treg cells and its function in MPE progression, providing novel insights into the epigenetic regulation of tumor development and metabolic strategies for MPE treatment by targeting lactate metabolism in Treg cells.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11683941/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142894411","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
High peripheral T cell diversity is associated with lower risk of toxicity and superior response to dual immune checkpoint inhibitor therapy in patients with metastatic NSCLC. 在转移性非小细胞肺癌患者中,高外周T细胞多样性与较低的毒性风险和对双重免疫检查点抑制剂治疗的优越反应相关。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-25 DOI: 10.1136/jitc-2024-008950
Mehmet Altan, Ruoxing Li, Ziyi Li, Runzhe Chen, Ajay Sheshadri, Hai T Tran, Latasha Little, Joshua Baguley, Jefferson Sinson, Natalie Vokes, Saumil Gandhi, Mara B Antonoff, Stephen G Swisher, Greg Lizee, Alexandre Reuben, John V Heymach, Jianjun Zhang

Introduction: Despite significant successes, immune checkpoint blockade fails to achieve clinical responses in a significant proportion of patients, predictive markers for responses are imperfect and immune-related adverse events (irAEs) are unpredictable. We used T-cell receptor (TCR) sequencing to systematically analyze prospectively collected patient blood samples from a randomized clinical trial of dual immune checkpoint inhibitor therapy to evaluate changes in the T-cell repertoire and their association with response and irAEs.

Methods: Patients with immunotherapy-naïve metastatic non-small cell lung cancer (NSCLC) were treated with ipilimumab and nivolumab according to trial protocol (LONESTAR, NCT03391869). Blood samples were systematically obtained at baseline (n=107), after 12 weeks of ipilimumab and nivolumab (n=91), and at the time of grade ≥2 irAEs (n=77). For analysis of T-cell repertoire, we performed immunoSEQ to assess the complementary determining region 3β region of the TCR involved in antigen binding.

Results: A total of 250 samples from 119 patients were analyzed. Patients who had a response to therapy exhibited greater T-cell diversity at baseline. Interestingly, patients with irAEs demonstrated lower T-cell richness at the time of toxicity compared with those without irAEs.

Conclusion: Our study highlights the potential impact of peripheral blood T-cell repertoire on clinical response and toxicities from the combination of ipilimumab and nivolumab in patients with metastatic NSCLC. These findings suggest that analysis of peripheral blood T-cell repertoire may help to guide patient selection for treatment with ipilimumab and nivolumab.

Trial registration number: NCT03391869.

尽管取得了显著的成功,但免疫检查点封锁在很大一部分患者中未能实现临床反应,反应的预测标记物不完善,免疫相关不良事件(irAEs)是不可预测的。我们使用t细胞受体(TCR)测序系统地分析了从双免疫检查点抑制剂治疗的随机临床试验中收集的前瞻性患者血液样本,以评估t细胞库的变化及其与反应和irAEs的关系。方法:根据试验方案(LONESTAR, NCT03391869),对immunotherapy-naïve转移性非小细胞肺癌(NSCLC)患者采用ipilimumab和nivolumab治疗。在基线(n=107)、ipilimumab和nivolumab治疗12周后(n=91)和irAEs≥2级时(n=77)系统采集血液样本。为了分析t细胞库,我们进行了免疫序列测序(immunoSEQ)来评估TCR中参与抗原结合的互补决定区3β区。结果:共分析119例患者250份样本。对治疗有反应的患者在基线时表现出更大的t细胞多样性。有趣的是,与没有irAEs的患者相比,irAEs患者在毒性发生时表现出较低的t细胞丰富度。结论:我们的研究强调了外周血t细胞库对转移性NSCLC患者伊匹单抗和纳武单抗联合治疗的临床反应和毒性的潜在影响。这些发现表明,分析外周血t细胞库可能有助于指导患者选择伊匹单抗和纳武单抗治疗。试验注册号:NCT03391869。
{"title":"High peripheral T cell diversity is associated with lower risk of toxicity and superior response to dual immune checkpoint inhibitor therapy in patients with metastatic NSCLC.","authors":"Mehmet Altan, Ruoxing Li, Ziyi Li, Runzhe Chen, Ajay Sheshadri, Hai T Tran, Latasha Little, Joshua Baguley, Jefferson Sinson, Natalie Vokes, Saumil Gandhi, Mara B Antonoff, Stephen G Swisher, Greg Lizee, Alexandre Reuben, John V Heymach, Jianjun Zhang","doi":"10.1136/jitc-2024-008950","DOIUrl":"10.1136/jitc-2024-008950","url":null,"abstract":"<p><strong>Introduction: </strong>Despite significant successes, immune checkpoint blockade fails to achieve clinical responses in a significant proportion of patients, predictive markers for responses are imperfect and immune-related adverse events (irAEs) are unpredictable. We used T-cell receptor (TCR) sequencing to systematically analyze prospectively collected patient blood samples from a randomized clinical trial of dual immune checkpoint inhibitor therapy to evaluate changes in the T-cell repertoire and their association with response and irAEs.</p><p><strong>Methods: </strong>Patients with immunotherapy-naïve metastatic non-small cell lung cancer (NSCLC) were treated with ipilimumab and nivolumab according to trial protocol (LONESTAR, NCT03391869). Blood samples were systematically obtained at baseline (n=107), after 12 weeks of ipilimumab and nivolumab (n=91), and at the time of grade ≥2 irAEs (n=77). For analysis of T-cell repertoire, we performed immunoSEQ to assess the complementary determining region 3β region of the TCR involved in antigen binding.</p><p><strong>Results: </strong>A total of 250 samples from 119 patients were analyzed. Patients who had a response to therapy exhibited greater T-cell diversity at baseline. Interestingly, patients with irAEs demonstrated lower T-cell richness at the time of toxicity compared with those without irAEs.</p><p><strong>Conclusion: </strong>Our study highlights the potential impact of peripheral blood T-cell repertoire on clinical response and toxicities from the combination of ipilimumab and nivolumab in patients with metastatic NSCLC. These findings suggest that analysis of peripheral blood T-cell repertoire may help to guide patient selection for treatment with ipilimumab and nivolumab.</p><p><strong>Trial registration number: </strong>NCT03391869.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11683914/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142894406","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
Engineered GM-CSF polarizes protumorigenic tumor-associated macrophages to an antitumorigenic phenotype and potently synergizes with IL-12 immunotherapy. 工程化的GM-CSF将致瘤性肿瘤相关巨噬细胞极化为抗致瘤表型,并与IL-12免疫疗法有效协同作用。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-22 DOI: 10.1136/jitc-2024-009541
Seounghun Kang, Aslan Mansurov, Trevin Kurtanich, Hye Rin Chun, Anna J Slezak, Lisa R Volpatti, Kevin Chang, Thomas Wang, Aaron T Alpar, Kirsten C Refvik, O Isabella Hansen, Gustavo J Borjas, Brendan T K Berg, Ha-Na Shim, Kevin T Hultgren, Suzana Gomes, Yue Wang, Ani Solanki, Jun Ishihara, Melody A Swartz, Jeffrey A Hubbell

Background: The use of immune checkpoint inhibitors (CPIs) has become a dominant regimen in modern cancer therapy, however immune resistance induced by tumor-associated macrophages (TAMs) with immune suppressive and evasion properties limits responses. Therefore, the rational design of immune modulators that can control the immune suppressive properties of TAMs and polarize them, as well as dendritic cells (DCs), toward a more proinflammatory phenotype is a principal objective in cancer immunotherapy.

Methods: Here, using a protein engineering approach to enhance cytokine residence in the tumor microenvironment, we examined combined stimulation of the myeloid compartment via tumor stroma-binding granulocyte-macrophage colony-stimulating factor (GM-CSF) to enhance responses in both DCs and T cells via stroma-binding interleukin-12 (IL-12). We evaluated tumor responses at the levels of outcome, cellular responses, and cytokine responses in both the tumors and the tumor-draining lymph nodes. We further investigated the potentiation of DC response to IL-12 by GM-CSF stimulation ex vivo.

Results: Engineered GM-CSF restored an antitumorigenic tumor myeloid microenvironment otherwise suppressed by TAMs, while engineered IL-12 provided effector signals to T cells, thereby boosting both tumor-resident antitumor macrophage and CD8+ T cell populations. Furthermore, engineered GM-CSF potentiated DC response to IL-12, upregulating DC expression of IL-12 receptor and enhancing their expression of proinflammatory cytokines and chemokines on IL-12 exposure. This resulted in remarkable synergistic efficacy in multiple solid tumor models treated with the dual cytokine combination. The combination therapy also improved the efficacy of CPI in a CPI-resistant genetically-engineered melanoma model and exhibited synergistic antitumor efficacy in a pulmonary metastasis model.

Conclusion: Our strategy provides a rational design for combination immunotherapy targeting both myeloid and lymphoid compartments through complementary mechanisms.

背景:使用免疫检查点抑制剂(CPIs)已成为现代癌症治疗的主要方案,然而,具有免疫抑制和逃避特性的肿瘤相关巨噬细胞(tam)诱导的免疫抵抗限制了应答。因此,合理设计免疫调节剂,控制tam的免疫抑制特性,并使它们以及树突状细胞(dc)向更促炎的表型分化,是癌症免疫治疗的主要目标。方法:本研究采用蛋白质工程方法增强细胞因子在肿瘤微环境中的驻留,研究了通过肿瘤基质结合的粒细胞-巨噬细胞集落刺激因子(GM-CSF)联合刺激髓系室,通过基质结合的白细胞介素-12 (IL-12)增强DCs和T细胞的反应。我们在肿瘤和肿瘤引流淋巴结的结果、细胞反应和细胞因子反应水平上评估肿瘤反应。我们进一步研究了GM-CSF刺激在体外增强DC对IL-12的反应。结果:工程化的GM-CSF恢复了抗肿瘤髓系微环境,而工程化的IL-12则向T细胞提供效应信号,从而增加肿瘤驻留的抗肿瘤巨噬细胞和CD8+ T细胞群。此外,工程化的GM-CSF增强了DC对IL-12的反应,上调了DC对IL-12受体的表达,并增强了DC对IL-12暴露的促炎细胞因子和趋化因子的表达。这导致双细胞因子联合治疗多种实体瘤模型具有显著的协同效应。联合治疗还提高了CPI在抗CPI基因工程黑色素瘤模型中的疗效,并在肺转移模型中显示出协同抗肿瘤疗效。结论:我们的策略通过互补机制为髓细胞和淋巴细胞的联合免疫治疗提供了合理的设计。
{"title":"Engineered GM-CSF polarizes protumorigenic tumor-associated macrophages to an antitumorigenic phenotype and potently synergizes with IL-12 immunotherapy.","authors":"Seounghun Kang, Aslan Mansurov, Trevin Kurtanich, Hye Rin Chun, Anna J Slezak, Lisa R Volpatti, Kevin Chang, Thomas Wang, Aaron T Alpar, Kirsten C Refvik, O Isabella Hansen, Gustavo J Borjas, Brendan T K Berg, Ha-Na Shim, Kevin T Hultgren, Suzana Gomes, Yue Wang, Ani Solanki, Jun Ishihara, Melody A Swartz, Jeffrey A Hubbell","doi":"10.1136/jitc-2024-009541","DOIUrl":"10.1136/jitc-2024-009541","url":null,"abstract":"<p><strong>Background: </strong>The use of immune checkpoint inhibitors (CPIs) has become a dominant regimen in modern cancer therapy, however immune resistance induced by tumor-associated macrophages (TAMs) with immune suppressive and evasion properties limits responses. Therefore, the rational design of immune modulators that can control the immune suppressive properties of TAMs and polarize them, as well as dendritic cells (DCs), toward a more proinflammatory phenotype is a principal objective in cancer immunotherapy.</p><p><strong>Methods: </strong>Here, using a protein engineering approach to enhance cytokine residence in the tumor microenvironment, we examined combined stimulation of the myeloid compartment via tumor stroma-binding granulocyte-macrophage colony-stimulating factor (GM-CSF) to enhance responses in both DCs and T cells via stroma-binding interleukin-12 (IL-12). We evaluated tumor responses at the levels of outcome, cellular responses, and cytokine responses in both the tumors and the tumor-draining lymph nodes. We further investigated the potentiation of DC response to IL-12 by GM-CSF stimulation ex vivo.</p><p><strong>Results: </strong>Engineered GM-CSF restored an antitumorigenic tumor myeloid microenvironment otherwise suppressed by TAMs, while engineered IL-12 provided effector signals to T cells, thereby boosting both tumor-resident antitumor macrophage and CD8<sup>+</sup> T cell populations. Furthermore, engineered GM-CSF potentiated DC response to IL-12, upregulating DC expression of IL-12 receptor and enhancing their expression of proinflammatory cytokines and chemokines on IL-12 exposure. This resulted in remarkable synergistic efficacy in multiple solid tumor models treated with the dual cytokine combination. The combination therapy also improved the efficacy of CPI in a CPI-resistant genetically-engineered melanoma model and exhibited synergistic antitumor efficacy in a pulmonary metastasis model.</p><p><strong>Conclusion: </strong>Our strategy provides a rational design for combination immunotherapy targeting both myeloid and lymphoid compartments through complementary mechanisms.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11667343/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142965096","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
Krebs von den Lungen-6 surveillance in immune checkpoint inhibitor-induced pneumonitis. 免疫检查点抑制剂诱导的肺炎中克雷布斯·冯·登·伦根-6的监测。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-22 DOI: 10.1136/jitc-2024-010114
Ryosuke Matsukane, Shoji Nakamura, Haruna Minami, Kazuya Tsubouchi, Yasuto Yoneshima, Kojiro Hata, Sai Yasukochi, Kimitaka Suetsugu, Isamu Okamoto, Takeshi Hirota

Background: The immune-related adverse event (irAE), pneumonitis, is a potentially fatal complication of immune checkpoint inhibitors (ICIs). Preventing its progression is crucial, emphasizing the need for effective screening tests. We evaluated the feasibility of using Krebs von den Lungen-6 (KL-6), a marker for interstitial pneumonitis, as a screening tool for pneumonitis.

Methods: We examined 500 patients with cancer divided into two groups: those with cancer other than non-small cell lung cancer (NSCLC) (Group 1, n=382) and those with NSCLC (Group 2, n=118). KL-6 levels were monitored before and during ICI treatment and analyzed for their correlation with pneumonitis.

Results: In Group 1, 37 patients (9.7%) developed pneumonitis. KL-6 levels were significantly elevated at irAE onset (pre: 222.0 U/mL, post: 743.0 U/mL, p<0.0001). Receiver operating characteristic curve analysis showed an area under the curve (AUC) of 0.903 (sensitivity 81.1%, specificity 91.6%) with a cut-off value 1.52 times pre-KL-6 levels, indicating that KL-6 is a reliable biomarker for pneumonitis. In these patients, the KL-6 level increased regardless of pneumonitis severity and was significantly elevated in patients with both symptomatic (pre: 205.0 U/mL, post: 674.5 U/mL, p<0.0001) and asymptomatic pneumonitis (pre: 314.0 U/mL, post: 743.0 U/mL, p<0.0001) at irAE onset. After irAE treatment, KL-6 levels in steroid-responsive patients remained unchanged; however, steroid-unresponsive patients had a significant increase in KL-6 levels at 1 month (1078 U/mL, p=0.031) compared with at irAE onset (678.0 U/mL). In Group 2, 24 patients (20.3%) developed irAE pneumonitis, with KL-6 levels elevated (pre: 360.5 U/mL, post: 506.5 U/mL, p=0.029) and an AUC of 0.683, indicating that KL-6 was less reliable in patients with NSCLC.

Conclusions: KL-6 is a viable screening biomarker in ICI-induced pneumonitis, particularly in patients without NSCLC. In patients with NSCLC, the significance of KL-6 monitoring is limited as it is not effective for detecting ICI-induced pneumonitis; their treatment is typically managed by pulmonary specialists. Early detection through KL-6 monitoring facilitates timely intervention for ICI-induced pneumonitis, potentially preventing treatment interruptions and reducing the need for immunosuppressants.

背景:免疫相关不良事件(irAE),肺炎,是免疫检查点抑制剂(ICIs)潜在的致命并发症。预防其发展至关重要,强调需要进行有效的筛查试验。我们评估了使用间质性肺炎标志物Krebs von den Lungen-6 (KL-6)作为肺炎筛查工具的可行性。方法:500例非小细胞肺癌(NSCLC)患者分为非小细胞肺癌组(1组,n=382)和非小细胞肺癌组(2组,n=118)。在ICI治疗前和治疗期间监测KL-6水平,并分析其与肺炎的相关性。结果:第1组37例(9.7%)发生肺炎。在irAE发病时,KL-6水平显著升高(发病前:222.0 U/mL,发病后:743.0 U/mL)。结论:KL-6是一种可行的筛查性生物标志物,尤其是在非小细胞肺癌患者中。在非小细胞肺癌患者中,KL-6监测的意义有限,因为它不能有效地检测ici引起的肺炎;他们的治疗通常由肺病专家管理。通过监测KL-6的早期发现有助于及时干预ici引起的肺炎,有可能防止治疗中断并减少对免疫抑制剂的需求。
{"title":"Krebs von den Lungen-6 surveillance in immune checkpoint inhibitor-induced pneumonitis.","authors":"Ryosuke Matsukane, Shoji Nakamura, Haruna Minami, Kazuya Tsubouchi, Yasuto Yoneshima, Kojiro Hata, Sai Yasukochi, Kimitaka Suetsugu, Isamu Okamoto, Takeshi Hirota","doi":"10.1136/jitc-2024-010114","DOIUrl":"10.1136/jitc-2024-010114","url":null,"abstract":"<p><strong>Background: </strong>The immune-related adverse event (irAE), pneumonitis, is a potentially fatal complication of immune checkpoint inhibitors (ICIs). Preventing its progression is crucial, emphasizing the need for effective screening tests. We evaluated the feasibility of using Krebs von den Lungen-6 (KL-6), a marker for interstitial pneumonitis, as a screening tool for pneumonitis.</p><p><strong>Methods: </strong>We examined 500 patients with cancer divided into two groups: those with cancer other than non-small cell lung cancer (NSCLC) (Group 1, n=382) and those with NSCLC (Group 2, n=118). KL-6 levels were monitored before and during ICI treatment and analyzed for their correlation with pneumonitis.</p><p><strong>Results: </strong>In Group 1, 37 patients (9.7%) developed pneumonitis. KL-6 levels were significantly elevated at irAE onset (pre: 222.0 U/mL, post: 743.0 U/mL, p<0.0001). Receiver operating characteristic curve analysis showed an area under the curve (AUC) of 0.903 (sensitivity 81.1%, specificity 91.6%) with a cut-off value 1.52 times pre-KL-6 levels, indicating that KL-6 is a reliable biomarker for pneumonitis. In these patients, the KL-6 level increased regardless of pneumonitis severity and was significantly elevated in patients with both symptomatic (pre: 205.0 U/mL, post: 674.5 U/mL, p<0.0001) and asymptomatic pneumonitis (pre: 314.0 U/mL, post: 743.0 U/mL, p<0.0001) at irAE onset. After irAE treatment, KL-6 levels in steroid-responsive patients remained unchanged; however, steroid-unresponsive patients had a significant increase in KL-6 levels at 1 month (1078 U/mL, p=0.031) compared with at irAE onset (678.0 U/mL). In Group 2, 24 patients (20.3%) developed irAE pneumonitis, with KL-6 levels elevated (pre: 360.5 U/mL, post: 506.5 U/mL, p=0.029) and an AUC of 0.683, indicating that KL-6 was less reliable in patients with NSCLC.</p><p><strong>Conclusions: </strong>KL-6 is a viable screening biomarker in ICI-induced pneumonitis, particularly in patients without NSCLC. In patients with NSCLC, the significance of KL-6 monitoring is limited as it is not effective for detecting ICI-induced pneumonitis; their treatment is typically managed by pulmonary specialists. Early detection through KL-6 monitoring facilitates timely intervention for ICI-induced pneumonitis, potentially preventing treatment interruptions and reducing the need for immunosuppressants.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11664336/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142965100","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
GrB-Fc-KS49, an anti-EMP2 granzyme B fusion protein therapeutic alters immune cell infiltration and suppresses breast cancer growth. 抗emp2颗粒酶B融合蛋白GrB-Fc-KS49治疗性改变免疫细胞浸润并抑制乳腺癌生长。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-22 DOI: 10.1136/jitc-2024-008891
Khalid A Mohamedali, Brian Aguirre, Cheng-Hsiang Lu, Anubhav Chandla, Nidhi Kejriwal, Lucia Liu, Ann M Chan, Lawrence H Cheung, SuYin Kok, Sergio Duarte, Ana Alvarez de Cienfuegos, David Casero, Michael G Rosenblum, Madhuri Wadehra

Background: Granzyme B (GrB) is a key effector molecule, delivered by cytotoxic T lymphocytes and natural killer cells during immune surveillance to induce cell death. Fusion proteins and immunoconjugates represent an innovative therapeutic approach to specifically deliver a deadly payload to target cells. Epithelial membrane protein-2 (EMP2) is highly expressed in invasive breast cancer (BC), including triple-negative BC (TNBC), and represents an attractive therapeutic target.

Methods: We designed a novel fusion protein (GrB-Fc-KS49) composed of an active GrB fused to an anti-EMP2 single-chain antibody tethered through the immunoglobulin G heavy chain (Fc) domain. We assessed the construct's GrB enzymatic activity, anti-EMP2 binding affinity, and cytotoxicity against a panel of BC cells. The construct's pharmacokinetics (PK), toxicity profile, and in vivo efficacy were also evaluated.

Results: GrB-Fc-KS49 exhibited comparable GrB enzymatic activity to commercial GrB, as well as high affinity to an EMP2 peptide, with the dissociation constant in the picomolar range. The fusion protein rapidly internalized into EMP2+cancer cells and showed in vitro cytotoxicity to cell lines expressing surface EMP2, with half-maximal cytotoxicity (IC50) values below 100 nM for most positive lines. Ex vivo stability at 37°C indicated a half-life exceeding 96 hours while in vivo PK indicated a biexponential plasma clearance, with a moderate initial clearance (t1/2α=18.4 hours) and a much slower terminal clearance rate (t1/2β=73.1 hours). No toxicity was measured in a Chem16 panel between the control and the GrB-Fc-KS49. In vivo, the GrB-Fc-KS49 showed efficacy against a TNBC syngeneic (4T1/FLuc) mouse model, reducing tumor volume and cell proliferation and increasing cell death compared with controls. Treatment using an EMT6 mouse model confirmed these results. In addition to a significant impact on cell proliferation, GrB-Fc-KS49 treatment also resulted in a dramatic increase of tumor-infiltrating CD45+ cells and redistribution of tumor-associated macrophages. Transcriptomic analysis of tumors post-treatment confirmed the remodeling of the immune tumor microenvironment by the GrB-Fc-KS49 immunotoxin.

Conclusions: GrB-Fc-KS49 showed high specificity and cytotoxicity towards EMP2-positive cells. In vivo, it reduced tumor burden and increased the recruitment of immune cells into the tumor, suggesting that GrB-Fc-KS49 is a promising therapeutic candidate against BC.

背景:颗粒酶B (Granzyme B, GrB)是细胞毒性T淋巴细胞和自然杀伤细胞在免疫监视过程中诱导细胞死亡的关键效应分子。融合蛋白和免疫偶联物代表了一种创新的治疗方法,可以特异性地向靶细胞递送致命的有效载荷。上皮膜蛋白-2 (EMP2)在浸润性乳腺癌(BC)中高表达,包括三阴性乳腺癌(TNBC),是一个有吸引力的治疗靶点。方法:设计了一种新的融合蛋白(GrB-Fc- ks49),该蛋白由活性GrB与通过免疫球蛋白G重链(Fc)结构域连接的抗emp2单链抗体融合而成。我们评估了构建物的GrB酶活性、抗emp2结合亲和力和对一组BC细胞的细胞毒性。该构建体的药代动力学(PK)、毒性特征和体内功效也进行了评估。结果:GrB- fc - ks49具有与商业GrB相当的GrB酶活性,并且与EMP2肽具有高亲和力,解离常数在皮摩尔范围内。融合蛋白迅速内化到EMP2阳性癌细胞中,对表面表达EMP2的细胞系显示出体外细胞毒性,大多数阳性细胞系的半数细胞毒性(IC50)值在100 nM以下。37°C的体外稳定性表明半衰期超过96小时,而体内PK显示双指数血浆清除率,初始清除率中等(t1/2α=18.4小时),最终清除率较慢(t1/2β=73.1小时)。在对照和GrB-Fc-KS49之间的Chem16面板中未检测到毒性。在体内,与对照组相比,GrB-Fc-KS49对TNBC同源(4T1/FLuc)小鼠模型有效,减少肿瘤体积和细胞增殖,增加细胞死亡。使用EMT6小鼠模型治疗证实了这些结果。除了对细胞增殖有显著影响外,GrB-Fc-KS49处理还导致肿瘤浸润性CD45+细胞的急剧增加和肿瘤相关巨噬细胞的重新分布。肿瘤治疗后的转录组学分析证实了GrB-Fc-KS49免疫毒素对肿瘤免疫微环境的重塑。结论:GrB-Fc-KS49对emp2阳性细胞具有较高的特异性和细胞毒性。在体内,它减少了肿瘤负荷,增加了免疫细胞进入肿瘤的募集,这表明GrB-Fc-KS49是一种有希望的治疗BC的候选药物。
{"title":"GrB-Fc-KS49, an anti-EMP2 granzyme B fusion protein therapeutic alters immune cell infiltration and suppresses breast cancer growth.","authors":"Khalid A Mohamedali, Brian Aguirre, Cheng-Hsiang Lu, Anubhav Chandla, Nidhi Kejriwal, Lucia Liu, Ann M Chan, Lawrence H Cheung, SuYin Kok, Sergio Duarte, Ana Alvarez de Cienfuegos, David Casero, Michael G Rosenblum, Madhuri Wadehra","doi":"10.1136/jitc-2024-008891","DOIUrl":"10.1136/jitc-2024-008891","url":null,"abstract":"<p><strong>Background: </strong>Granzyme B (GrB) is a key effector molecule, delivered by cytotoxic T lymphocytes and natural killer cells during immune surveillance to induce cell death. Fusion proteins and immunoconjugates represent an innovative therapeutic approach to specifically deliver a deadly payload to target cells. Epithelial membrane protein-2 (EMP2) is highly expressed in invasive breast cancer (BC), including triple-negative BC (TNBC), and represents an attractive therapeutic target.</p><p><strong>Methods: </strong>We designed a novel fusion protein (GrB-Fc-KS49) composed of an active GrB fused to an anti-EMP2 single-chain antibody tethered through the immunoglobulin G heavy chain (Fc) domain. We assessed the construct's GrB enzymatic activity, anti-EMP2 binding affinity, and cytotoxicity against a panel of BC cells. The construct's pharmacokinetics (PK), toxicity profile, and in vivo efficacy were also evaluated.</p><p><strong>Results: </strong>GrB-Fc-KS49 exhibited comparable GrB enzymatic activity to commercial GrB, as well as high affinity to an EMP2 peptide, with the dissociation constant in the picomolar range. The fusion protein rapidly internalized into EMP2+cancer cells and showed in vitro cytotoxicity to cell lines expressing surface EMP2, with half-maximal cytotoxicity (IC<sub>50</sub>) values below 100 nM for most positive lines. Ex vivo stability at 37°C indicated a half-life exceeding 96 hours while in vivo PK indicated a biexponential plasma clearance, with a moderate initial clearance (t<sub>1/2</sub>α=18.4 hours) and a much slower terminal clearance rate (t<sub>1/2</sub>β=73.1 hours). No toxicity was measured in a Chem16 panel between the control and the GrB-Fc-KS49. In vivo, the GrB-Fc-KS49 showed efficacy against a TNBC syngeneic (4T1/<sub>FLuc</sub>) mouse model, reducing tumor volume and cell proliferation and increasing cell death compared with controls. Treatment using an EMT6 mouse model confirmed these results. In addition to a significant impact on cell proliferation, GrB-Fc-KS49 treatment also resulted in a dramatic increase of tumor-infiltrating CD45+ cells and redistribution of tumor-associated macrophages. Transcriptomic analysis of tumors post-treatment confirmed the remodeling of the immune tumor microenvironment by the GrB-Fc-KS49 immunotoxin.</p><p><strong>Conclusions: </strong>GrB-Fc-KS49 showed high specificity and cytotoxicity towards EMP2-positive cells. In vivo, it reduced tumor burden and increased the recruitment of immune cells into the tumor, suggesting that GrB-Fc-KS49 is a promising therapeutic candidate against BC.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11667298/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142965098","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
Neuropsychiatric manifestations following chimeric antigen receptor T cell therapy for cancer: a systematic review of clinical outcomes and management strategies. 嵌合抗原受体T细胞治疗癌症后的神经精神表现:临床结果和管理策略的系统回顾。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-22 DOI: 10.1136/jitc-2024-009174
Anna Fleischer, Sophia Kurth, Johannes Duell, Max Topp, Patrick-Pascal Strunz, Julia Mersi, Leo Rasche, Carmen Sanges, Michael Hudecek, Hermann Einsele, Imad Maatouk

Background: Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative modality in the treatment of patients with cancer. However, it is increasingly evident that this therapeutic approach is not without its challenges. The unique nature of CAR-T cells as living drugs introduces a distinct set of side effects. As the application of CAR-T cell therapy expands to treat a broader range of diseases, it becomes increasingly important to devise effective strategies for handling the associated toxicities. Challenges in treating patients with CAR-T cells include addressing complications such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and cytopenias. This comprehensive review seeks to systematically identify, categorize and elucidate all previously described neurological and psychological side effects associated with CAR-T cell therapy, shedding light on the pertinent laboratory findings that underscore these phenomena.

Methods: PubMed, Springer Link, and ScienceDirect were systematically searched for empirical studies on adult patients with cancer receiving CAR-T cell therapy for hemato-oncological malignancies. Quality assessment was conducted using Version 2 of the Cochrane risk-of-bias tool (RoB 2) for randomized trials and adherence to the STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) checklist for observational studies. The synthesis of findings was conducted via a narrative approach, consolidating the diverse array of data into a coherent framework.

Results: From an initial pool of 2,276 citations, 546 studies met the inclusion criteria, exhibiting a rich tapestry of heterogeneity in terms of study characteristics and patient samples. The incidence of neuropsychological symptoms varied notably across different CAR-T cell products and hematological malignancies. Among the most frequently reported neuropsychological symptoms were aphasia, attention deficits, impaired consciousness, and disorientation, alongside a constellation of other symptoms including confusion, cognitive impairment, memory loss, writing difficulties, fatigue, headache, agitation, tremor, seizures, and psychomotor retardation. Early intervention strategies, including corticosteroids and tocilizumab, have shown the potential to reduce the intensity of neuropsychological symptoms and prevent their progression to critical complications.

Conclusion: These insights underscore the imperative of extending neuropsychological assessments beyond the conventional Immune Effector Cell-Associated Encephalopathy score framework.

背景:嵌合抗原受体(CAR)-T细胞疗法已成为癌症患者治疗的一种变革性方式。然而,越来越明显的是,这种治疗方法并非没有挑战。CAR-T细胞作为活药物的独特性质带来了一系列独特的副作用。随着CAR-T细胞疗法的应用扩展到治疗更广泛的疾病,设计有效的策略来处理相关的毒性变得越来越重要。治疗CAR-T细胞患者的挑战包括解决诸如细胞因子释放综合征、免疫效应细胞相关神经毒性综合征和细胞减少症等并发症。这篇全面的综述旨在系统地识别、分类和阐明所有先前描述的与CAR-T细胞治疗相关的神经和心理副作用,揭示强调这些现象的相关实验室发现。方法:系统检索PubMed、施普林格Link和ScienceDirect,检索成人癌症患者接受CAR-T细胞治疗血液肿瘤恶性肿瘤的实证研究。使用Cochrane第2版偏倚风险工具(RoB 2)对随机试验进行质量评估,并对观察性研究遵守STROBE(加强流行病学观察性研究报告)检查表。调查结果的综合是通过叙述方法进行的,将各种各样的数据整合到一个连贯的框架中。结果:从最初的2276个引用中,546个研究符合纳入标准,在研究特征和患者样本方面表现出丰富的异质性。神经心理症状的发生率在不同的CAR-T细胞产物和血液系统恶性肿瘤之间有显著差异。最常见的神经心理学症状包括失语、注意力缺陷、意识受损和定向障碍,以及一系列其他症状,包括精神错乱、认知障碍、记忆丧失、书写困难、疲劳、头痛、躁动、震颤、癫痫发作和精神运动迟缓。包括皮质类固醇和托珠单抗在内的早期干预策略已显示出降低神经心理症状强度并防止其发展为严重并发症的潜力。结论:这些见解强调了在传统的免疫效应细胞相关脑病评分框架之外扩展神经心理学评估的必要性。
{"title":"Neuropsychiatric manifestations following chimeric antigen receptor T cell therapy for cancer: a systematic review of clinical outcomes and management strategies.","authors":"Anna Fleischer, Sophia Kurth, Johannes Duell, Max Topp, Patrick-Pascal Strunz, Julia Mersi, Leo Rasche, Carmen Sanges, Michael Hudecek, Hermann Einsele, Imad Maatouk","doi":"10.1136/jitc-2024-009174","DOIUrl":"10.1136/jitc-2024-009174","url":null,"abstract":"<p><strong>Background: </strong>Chimeric antigen receptor (CAR)-T cell therapy has emerged as a transformative modality in the treatment of patients with cancer. However, it is increasingly evident that this therapeutic approach is not without its challenges. The unique nature of CAR-T cells as living drugs introduces a distinct set of side effects. As the application of CAR-T cell therapy expands to treat a broader range of diseases, it becomes increasingly important to devise effective strategies for handling the associated toxicities. Challenges in treating patients with CAR-T cells include addressing complications such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and cytopenias. This comprehensive review seeks to systematically identify, categorize and elucidate all previously described neurological and psychological side effects associated with CAR-T cell therapy, shedding light on the pertinent laboratory findings that underscore these phenomena.</p><p><strong>Methods: </strong>PubMed, Springer Link, and ScienceDirect were systematically searched for empirical studies on adult patients with cancer receiving CAR-T cell therapy for hemato-oncological malignancies. Quality assessment was conducted using Version 2 of the Cochrane risk-of-bias tool (RoB 2) for randomized trials and adherence to the STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) checklist for observational studies. The synthesis of findings was conducted via a narrative approach, consolidating the diverse array of data into a coherent framework.</p><p><strong>Results: </strong>From an initial pool of 2,276 citations, 546 studies met the inclusion criteria, exhibiting a rich tapestry of heterogeneity in terms of study characteristics and patient samples. The incidence of neuropsychological symptoms varied notably across different CAR-T cell products and hematological malignancies. Among the most frequently reported neuropsychological symptoms were aphasia, attention deficits, impaired consciousness, and disorientation, alongside a constellation of other symptoms including confusion, cognitive impairment, memory loss, writing difficulties, fatigue, headache, agitation, tremor, seizures, and psychomotor retardation. Early intervention strategies, including corticosteroids and tocilizumab, have shown the potential to reduce the intensity of neuropsychological symptoms and prevent their progression to critical complications.</p><p><strong>Conclusion: </strong>These insights underscore the imperative of extending neuropsychological assessments beyond the conventional Immune Effector Cell-Associated Encephalopathy score framework.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11667355/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142965102","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
Preclinical model for evaluating human TCRs against chimeric syngeneic tumors. 评估人tcr抗嵌合同基因肿瘤的临床前模型。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-12-22 DOI: 10.1136/jitc-2024-009504
Aikaterini Semilietof, Evangelos Stefanidis, Elise Gray-Gaillard, Julien Pujol, Alessia D'Esposito, Patrick Reichenbach, Philippe Guillaume, Vincent Zoete, Melita Irving, Olivier Michielin

Background: The adoptive cell transfer (ACT) of T cell receptor (TCR)-engineered T cells targeting the HLA-A2-restricted epitope NY-ESO-1157-165 (A2/NY) has yielded important clinical responses against several cancers. A variety of approaches are being taken to augment tumor control by ACT including TCR affinity-optimization and T-cell coengineering strategies to address the suppressive tumor microenvironment (TME). Most TCRs of clinical interest are evaluated in immunocompromised mice to enable human T-cell engraftment and do not recapitulate the dynamic interplay that occurs with endogenous immunity in a treated patient. A variety of humanized mouse models have been described but they have limitations in immune reconstitution and are technically challenging to implement. Here, we have developed a chimeric syngeneic tumor model in which A2Kb transgenic C57BL/6 mice are engrafted with B16 expressing A2Kb:NY as a single chain trimer (SCT) and treated by ACT with murine T cells expressing A2/NY TCRs comprising human variable fused to mouse constant regions.

Methods: We compared the function of a supraphysiological affinity A2/NY TCR (wtc51m), a computationally designed TCR in an optimal affinity range (DMβ), and a near non-binding TCR (V49I), engineered in both primary human and murine T cells by lentiviral and retroviral transduction, respectively. We evaluated a variety of strategies to stably express A2Kb:NY on the surface of mouse tumor cell lines including B16 melanoma, ultimately achieving success with an SCT comprising human β2m fused by GS linkers to both the NY-peptide and to α1 of the HLA complex. ACT studies were performed in B16-A2Kb:NY tumor-bearing, non-preconditioned immune-competent HLA-A*0201/H-2Kb (A2Kb) transgenic C57BL/6 mice and tumors characterized post-transfer.

Results: We observed significantly improved function of DMβ-T cells as well as superior infiltration and tumor control upon ACT as compared to the control TCR-T cells. Moreover, with our chimeric syngeneic tumor model, we were able to track dynamic and favorable changes in the TME upon DMβ-T cell transfer.

Conclusions: We have developed a robust, simple, and inexpensive preclinical strategy for evaluating human TCRs in the context of a fully competent murine immune system that can aid in the development of coengineered TCR-T cells and combination treatments translated to the clinic.

背景:T细胞受体(TCR)工程T细胞靶向hla -A2限制性表位NY- eso -1157-165 (A2/NY)的过继细胞转移(ACT)已经对几种癌症产生了重要的临床反应。人们正在采取多种方法来增强ACT对肿瘤的控制,包括TCR亲和优化和t细胞协同工程策略,以解决抑制性肿瘤微环境(TME)。大多数具有临床意义的tcr都是在免疫功能低下的小鼠中进行评估,以使人类t细胞植入,并且不能重现治疗患者体内与内源性免疫发生的动态相互作用。各种各样的人源化小鼠模型已经被描述,但它们在免疫重建方面有局限性,并且在技术上具有挑战性。在这里,我们建立了一个嵌合的同基因肿瘤模型,将A2Kb转基因C57BL/6小鼠植入表达A2Kb:NY的B16作为单链三聚体(SCT),并用ACT处理表达A2/NY TCRs的小鼠T细胞,其中包含人类变量融合到小鼠恒定区域。方法:我们比较了A2/NY TCR (wtc51m)、计算设计的最佳亲和力范围TCR (DMβ)和近非结合TCR (V49I)的功能,它们分别通过慢病毒和逆转录病毒转导在人和小鼠原代T细胞中进行工程设计。我们评估了多种在小鼠肿瘤细胞系(包括B16黑色素瘤)表面稳定表达A2Kb:NY的策略,最终成功地将人β2m通过GS连接体融合到NY肽和HLA复合物的α1上。ACT研究在B16-A2Kb:NY荷瘤、非预处理免疫活性HLA-A*0201/H-2Kb (A2Kb)转基因C57BL/6小鼠和转移后的肿瘤中进行。结果:与对照TCR-T细胞相比,ACT对DMβ-T细胞的功能有明显改善,浸润和肿瘤控制也有明显改善。此外,利用我们的嵌合同基因肿瘤模型,我们能够跟踪DMβ-T细胞转移时TME的动态和有利变化。结论:我们已经开发出一种强大、简单、廉价的临床前策略,用于在完全胜任的小鼠免疫系统背景下评估人类tcr,可以帮助开发协同工程TCR-T细胞和转化为临床的联合治疗。
{"title":"Preclinical model for evaluating human TCRs against chimeric syngeneic tumors.","authors":"Aikaterini Semilietof, Evangelos Stefanidis, Elise Gray-Gaillard, Julien Pujol, Alessia D'Esposito, Patrick Reichenbach, Philippe Guillaume, Vincent Zoete, Melita Irving, Olivier Michielin","doi":"10.1136/jitc-2024-009504","DOIUrl":"10.1136/jitc-2024-009504","url":null,"abstract":"<p><strong>Background: </strong>The adoptive cell transfer (ACT) of T cell receptor (TCR)-engineered T cells targeting the HLA-A2-restricted epitope NY-ESO-1<sub>157-165</sub> (A2/NY) has yielded important clinical responses against several cancers. A variety of approaches are being taken to augment tumor control by ACT including TCR affinity-optimization and T-cell coengineering strategies to address the suppressive tumor microenvironment (TME). Most TCRs of clinical interest are evaluated in immunocompromised mice to enable human T-cell engraftment and do not recapitulate the dynamic interplay that occurs with endogenous immunity in a treated patient. A variety of humanized mouse models have been described but they have limitations in immune reconstitution and are technically challenging to implement. Here, we have developed a chimeric syngeneic tumor model in which A2Kb transgenic C57BL/6 mice are engrafted with B16 expressing A2Kb:NY as a single chain trimer (SCT) and treated by ACT with murine T cells expressing A2/NY TCRs comprising human variable fused to mouse constant regions.</p><p><strong>Methods: </strong>We compared the function of a supraphysiological affinity A2/NY TCR (wtc51m), a computationally designed TCR in an optimal affinity range (DMβ), and a near non-binding TCR (V49I), engineered in both primary human and murine T cells by lentiviral and retroviral transduction, respectively. We evaluated a variety of strategies to stably express A2Kb:NY on the surface of mouse tumor cell lines including B16 melanoma, ultimately achieving success with an SCT comprising human β2m fused by GS linkers to both the NY-peptide and to α1 of the HLA complex. ACT studies were performed in B16-A2Kb:NY tumor-bearing, non-preconditioned immune-competent HLA-A*0201/H-2Kb (A2Kb) transgenic C57BL/6 mice and tumors characterized post-transfer.</p><p><strong>Results: </strong>We observed significantly improved function of DMβ-T cells as well as superior infiltration and tumor control upon ACT as compared to the control TCR-T cells. Moreover, with our chimeric syngeneic tumor model, we were able to track dynamic and favorable changes in the TME upon DMβ-T cell transfer.</p><p><strong>Conclusions: </strong>We have developed a robust, simple, and inexpensive preclinical strategy for evaluating human TCRs in the context of a fully competent murine immune system that can aid in the development of coengineered TCR-T cells and combination treatments translated to the clinic.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 12","pages":""},"PeriodicalIF":10.3,"publicationDate":"2024-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11667476/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142965013","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal for Immunotherapy of Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
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