首页 > 最新文献

Journal for Immunotherapy of Cancer最新文献

英文 中文
CREB3L1 facilitates pancreatic tumor progression and reprograms intratumoral tumor-associated macrophages to shape an immunotherapy-resistance microenvironment. CREB3L1促进胰腺肿瘤进展并重新编程肿瘤内肿瘤相关巨噬细胞,形成免疫治疗抵抗微环境。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-06 DOI: 10.1136/jitc-2024-010029
Haiyan Xu, Shengbai Xue, Yang Sun, Jingyu Ma, Shumin Li, Yanling Wang, Tiebo Mao, Weiyu Ge, Ming Yue, Daiyuan Shentu, Wenxin Lu, Yongchao Wang, Jiong Hu, Jiujie Cui, Xiaofei Zhang, Li Cai, Yu Wang, Liwei Wang

Background: To date, a growing body of evidence suggests that unfolded protein response (UPR) sensors play a vital role in carcinogenesis. However, it remains unclear whether they are involved in pancreatic ductal adenocarcinoma (PDAC) and how they relate to clinical outcomes. This study aims to investigate the biological function and mechanism of how a novel UPR sensor, CREB3L1 works in PDAC and further evaluate its clinical application prospect.

Methods: We tested UPR signaling including CREB3L1 in Thapsigargin-treated PDAC cells. Subsequently, we defined CREB3L1 expression and further analyzed its expression with clinical characteristics in PDAC. Then, we established gene-modified cells to determine whether CREB3L1 functions in cell proliferation and migration capacity. Besides, we constructed subcutaneously and orthotopically transplanted mice models to verify their progrowing function and pulmonary metastasis models to prove their proinvasion role. What's more, RNAseq, qPCR, Western blotting, immunohistochemistry and multicolor flow cytometry experiments were used to explore the mechanism of how CREB3L1 worked in PDAC. Lastly, CREB3L1 expression correlation with PDAC immunotherapy outcome and immune cell signatures were explored in the patients with advanced PDAC who received PD-1 antibody therapy.

Results: We first confirmed CREB3L1 could be induced by endoplasmic reticulum stressor and found its aberrant activation was associated with poorer overall survival in PDAC patients indicating the protumor function of the new UPR sensor. Functionally, we confirmed CREB3L1 contributing to PDAC malignant progression including growth and metastasis by multiple in in vitro and in vivo models. Mechanistically, CREB3L1 upregulated COL3A1 and promoted dense stroma formation for facilitating PDAC and knocking down COL3A1 disrupted CREB3L1 protumor function. Furthermore, CREB3L1-induced TAM polarization toward an M2 phenotype and reduced the infiltration of CD8+ T cells. Clinically, CREB3L1 correlated with immune cell signatures as well as immune checkpoint blockade (ICB) treatment response and outcome that CREB3L1aberrant activation indicated poorer efficacy and worse prognosis than the low in PDAC which might empower clinical decision.

Conclusions: Collectively, this study revealed CREB3L1 facilitated PDAC progression, shaped an immune exclude tumor microenvironment and distinguished therapy response and outcome of ICB therapy indicating CREB3L1 could be a promising novel molecular target and biomarker for PDAC treatment.

背景:迄今为止,越来越多的证据表明未折叠蛋白反应(UPR)传感器在癌症发生中起着至关重要的作用。然而,目前尚不清楚它们是否与胰腺导管腺癌(PDAC)有关,以及它们与临床结果的关系。本研究旨在探讨新型UPR传感器CREB3L1在PDAC中的生物学功能和作用机制,并进一步评价其临床应用前景。方法:我们在thapsigargin处理的PDAC细胞中检测包括CREB3L1在内的UPR信号。随后,我们定义了CREB3L1的表达,并进一步分析了其在PDAC中的表达与临床特征。然后,我们建立了基因修饰的细胞,以确定CREB3L1是否在细胞增殖和迁移能力中起作用。此外,我们建立小鼠皮下和原位移植模型来验证其促生长功能,并建立肺转移模型来证明其预防侵袭作用。通过RNAseq、qPCR、Western blotting、免疫组织化学、多色流式细胞术等实验探讨CREB3L1在PDAC中的作用机制。最后,在接受PD-1抗体治疗的晚期PDAC患者中,探讨CREB3L1表达与PDAC免疫治疗结果和免疫细胞特征的相关性。结果:我们首次证实了CREB3L1可以被内质网应激源诱导,并发现其异常激活与PDAC患者较差的总生存率相关,这表明了新型UPR传感器的肿瘤功能。在功能上,我们通过多种体外和体内模型证实CREB3L1参与了PDAC的恶性进展,包括生长和转移。机制上,CREB3L1上调COL3A1,促进致密间质形成,促进PDAC,敲低COL3A1破坏CREB3L1蛋白功能。此外,creb3l1诱导TAM向M2表型极化,减少CD8+ T细胞的浸润。在临床上,CREB3L1与免疫细胞特征和免疫检查点阻断(ICB)治疗反应相关,与PDAC低水平相比,CREB3L1异常激活的疗效更差,预后更差,这可能有助于临床决策。结论:总的来说,本研究揭示了CREB3L1促进PDAC进展,形成免疫排斥肿瘤微环境,并区分ICB治疗的治疗反应和结果,表明CREB3L1可能是一个有希望的PDAC治疗的新分子靶点和生物标志物。
{"title":"CREB3L1 facilitates pancreatic tumor progression and reprograms intratumoral tumor-associated macrophages to shape an immunotherapy-resistance microenvironment.","authors":"Haiyan Xu, Shengbai Xue, Yang Sun, Jingyu Ma, Shumin Li, Yanling Wang, Tiebo Mao, Weiyu Ge, Ming Yue, Daiyuan Shentu, Wenxin Lu, Yongchao Wang, Jiong Hu, Jiujie Cui, Xiaofei Zhang, Li Cai, Yu Wang, Liwei Wang","doi":"10.1136/jitc-2024-010029","DOIUrl":"10.1136/jitc-2024-010029","url":null,"abstract":"<p><strong>Background: </strong>To date, a growing body of evidence suggests that unfolded protein response (UPR) sensors play a vital role in carcinogenesis. However, it remains unclear whether they are involved in pancreatic ductal adenocarcinoma (PDAC) and how they relate to clinical outcomes. This study aims to investigate the biological function and mechanism of how a novel UPR sensor, CREB3L1 works in PDAC and further evaluate its clinical application prospect.</p><p><strong>Methods: </strong>We tested UPR signaling including CREB3L1 in Thapsigargin-treated PDAC cells. Subsequently, we defined CREB3L1 expression and further analyzed its expression with clinical characteristics in PDAC. Then, we established gene-modified cells to determine whether CREB3L1 functions in cell proliferation and migration capacity. Besides, we constructed subcutaneously and orthotopically transplanted mice models to verify their progrowing function and pulmonary metastasis models to prove their proinvasion role. What's more, RNAseq, qPCR, Western blotting, immunohistochemistry and multicolor flow cytometry experiments were used to explore the mechanism of how CREB3L1 worked in PDAC. Lastly, CREB3L1 expression correlation with PDAC immunotherapy outcome and immune cell signatures were explored in the patients with advanced PDAC who received PD-1 antibody therapy.</p><p><strong>Results: </strong>We first confirmed CREB3L1 could be induced by endoplasmic reticulum stressor and found its aberrant activation was associated with poorer overall survival in PDAC patients indicating the protumor function of the new UPR sensor. Functionally, we confirmed CREB3L1 contributing to PDAC malignant progression including growth and metastasis by multiple in in vitro and in vivo models. Mechanistically, CREB3L1 upregulated COL3A1 and promoted dense stroma formation for facilitating PDAC and knocking down COL3A1 disrupted CREB3L1 protumor function. Furthermore, CREB3L1-induced TAM polarization toward an M2 phenotype and reduced the infiltration of CD8<sup>+</sup> T cells. Clinically, CREB3L1 correlated with immune cell signatures as well as immune checkpoint blockade (ICB) treatment response and outcome that CREB3L1aberrant activation indicated poorer efficacy and worse prognosis than the low in PDAC which might empower clinical decision.</p><p><strong>Conclusions: </strong>Collectively, this study revealed CREB3L1 facilitated PDAC progression, shaped an immune exclude tumor microenvironment and distinguished therapy response and outcome of ICB therapy indicating CREB3L1 could be a promising novel molecular target and biomarker for PDAC treatment.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749327/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143005594","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
XCL1-secreting CEA CAR-T cells enhance endogenous CD8+ T cell responses to tumor neoantigens to confer a long-term antitumor immunity. 分泌xcl1的CEA CAR-T细胞增强内源性CD8+ T细胞对肿瘤新抗原的反应,从而获得长期的抗肿瘤免疫。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-06 DOI: 10.1136/jitc-2024-010581
Xing-Ning Li, Feifei Wang, Kun Chen, Zhiyuan Wu, Ruochan Zhang, Chentong Xiao, Fei Zhao, Dongmei Wang, Hong Zhao, Yuliang Ran, Chunfeng Qu

Background: Therapeutic efficacy of carcinoembryonic antigen (CEA)-specific chimeric antigen receptor (CAR) T cells against colorectal cancer (CRC) remains limited due to the unique characteristics and distinct microenvironments of tumor tissues. We modified CEA-specific CAR-T cells, aiming to stimulate endogenous CD8+ T cell responses against neoantigens that were derived from CEA-positive tumors destroyed by the CAR T cells.

Methods: In a conventional CEA CAR (reg-CAR), we modified it to express lymphotactin XCL1 and interleukin (IL)-7 genes, constructing a modified 7XCL1-CAR. By generating the CEA-specific 7XCL1-CAR T cells, we assessed their antitumor efficacy against CRC cells with varying levels of CEA expression, both in cell-cultures and in two strains of tumor-bearing syngeneic mice.

Results: Following retroviral transduction, 7XCL1-CAR T cells and reg-CAR T cells exhibited similar positive proportions of CEA-CAR and CD4:CD8 ratios. In co-culture system with CEA-negative CT26 cells, no differences in cytotoxicity were observed between 7XCL1-CAR and reg-CAR T cells. However, in co-culture with CT26.CEAhigh and CT26.CEAint cells, 7XCL1-CAR T cells displayed higher cytotoxicity than that reg-CAR T cells after 60 hours. On interaction with CT26.CEA-positive cells, 7XCL1-CAR T cells secreted higher levels of XCL1 and IL-7, effectively recruited the most potent cross-presenting cDC1s (type-I conventional dendritic cells), and sustained the antitumor activity of CAR-T cells. In treating mice that carried tumors derived from universally CEA-positive cells, 7XCL1-CAR T cells exhibited no difference compared with reg-CAR T cells. However, in treating mice with tumors containing both CEA-positive and CEA-negative cells, 7XCL1-CAR T cells displayed greater inhibition than that of reg-CAR-T cells. After treatment of 7XCL1-CAR T cells, tumor-bearing mice exhibited enhanced infiltration of cDC1s, maintained CAR-T activity, and generation of endogenous neoantigen-specific T cells. Consequently, 7XCL1-CAR T cell-treated mice demonstrated resistance to challenge with CEA-negative CT26 cells.

Conclusion: Treatment with CEA-specific, XCL1-secreting CAR-T cells for CEA-positive tumors promoted the generation of CD8+ T cells against tumor neoantigens, mediating a long-term antitumor immunity against heterogeneous CRCs.

背景:癌胚抗原(CEA)特异性嵌合抗原受体(CAR) T细胞治疗结直肠癌(CRC)的疗效仍然有限,因为肿瘤组织的独特特性和不同的微环境。我们修饰了cea特异性CAR-T细胞,旨在刺激内源性CD8+ T细胞对来自被CAR-T细胞破坏的cea阳性肿瘤的新抗原的反应。方法:在常规CEA CAR (reg-CAR)中,对其进行修饰,表达淋巴趋动素XCL1和白细胞介素(IL)-7基因,构建修饰后的7XCL1-CAR。通过生成CEA特异性的7XCL1-CAR - T细胞,我们在细胞培养和两种携带肿瘤的同基因小鼠中评估了它们对不同CEA表达水平的CRC细胞的抗肿瘤功效。结果:逆转录病毒转导后,7XCL1-CAR - T细胞和reg-CAR - T细胞的CEA-CAR和CD4:CD8比值呈相似的阳性比例。在与cea阴性CT26细胞共培养的系统中,7XCL1-CAR和regg - car T细胞的细胞毒性没有差异。然而,与CT26共培养。CEAhigh和CT26。60小时后,7XCL1-CAR - T细胞比regi - car - T细胞表现出更高的细胞毒性。与CT26的相互作用。cea阳性细胞,7XCL1-CAR -T细胞分泌更高水平的XCL1和IL-7,有效募集最有效的交叉呈递cDC1s (i型常规树突状细胞),并维持CAR-T细胞的抗肿瘤活性。在治疗携带来自普遍cea阳性细胞的肿瘤的小鼠时,7XCL1-CAR - T细胞与regg - car - T细胞相比没有表现出差异。然而,在治疗含有cea阳性和cea阴性细胞的肿瘤小鼠时,7XCL1-CAR -T细胞比regg - car -T细胞表现出更大的抑制作用。经7XCL1-CAR -T细胞处理后,肿瘤小鼠表现出cDC1s浸润增强,CAR-T活性维持,内源性新抗原特异性T细胞生成。因此,7XCL1-CAR - T细胞处理的小鼠对cea阴性CT26细胞的攻击表现出抵抗性。结论:用cea特异性、分泌xcl1的CAR-T细胞治疗cea阳性肿瘤,促进了针对肿瘤新抗原的CD8+ T细胞的产生,介导了针对异质crc的长期抗肿瘤免疫。
{"title":"XCL1-secreting CEA CAR-T cells enhance endogenous CD8<sup>+</sup> T cell responses to tumor neoantigens to confer a long-term antitumor immunity.","authors":"Xing-Ning Li, Feifei Wang, Kun Chen, Zhiyuan Wu, Ruochan Zhang, Chentong Xiao, Fei Zhao, Dongmei Wang, Hong Zhao, Yuliang Ran, Chunfeng Qu","doi":"10.1136/jitc-2024-010581","DOIUrl":"10.1136/jitc-2024-010581","url":null,"abstract":"<p><strong>Background: </strong>Therapeutic efficacy of carcinoembryonic antigen (CEA)-specific chimeric antigen receptor (CAR) T cells against colorectal cancer (CRC) remains limited due to the unique characteristics and distinct microenvironments of tumor tissues. We modified CEA-specific CAR-T cells, aiming to stimulate endogenous CD8<sup>+</sup> T cell responses against neoantigens that were derived from CEA-positive tumors destroyed by the CAR T cells.</p><p><strong>Methods: </strong>In a conventional CEA CAR (reg-CAR), we modified it to express lymphotactin XCL1 and interleukin (IL)-7 genes, constructing a modified 7XCL1-CAR. By generating the CEA-specific 7XCL1-CAR T cells, we assessed their antitumor efficacy against CRC cells with varying levels of CEA expression, both in cell-cultures and in two strains of tumor-bearing syngeneic mice.</p><p><strong>Results: </strong>Following retroviral transduction, 7XCL1-CAR T cells and reg-CAR T cells exhibited similar positive proportions of CEA-CAR and CD4:CD8 ratios. In co-culture system with CEA-negative CT26 cells, no differences in cytotoxicity were observed between 7XCL1-CAR and reg-CAR T cells. However, in co-culture with CT26.CEA<sup>high</sup> and CT26.CEA<sup>int</sup> cells, 7XCL1-CAR T cells displayed higher cytotoxicity than that reg-CAR T cells after 60 hours. On interaction with CT26.CEA-positive cells, 7XCL1-CAR T cells secreted higher levels of XCL1 and IL-7, effectively recruited the most potent cross-presenting cDC1s (type-I conventional dendritic cells), and sustained the antitumor activity of CAR-T cells. In treating mice that carried tumors derived from universally CEA-positive cells, 7XCL1-CAR T cells exhibited no difference compared with reg-CAR T cells. However, in treating mice with tumors containing both CEA-positive and CEA-negative cells, 7XCL1-CAR T cells displayed greater inhibition than that of reg-CAR-T cells. After treatment of 7XCL1-CAR T cells, tumor-bearing mice exhibited enhanced infiltration of cDC1s, maintained CAR-T activity, and generation of endogenous neoantigen-specific T cells. Consequently, 7XCL1-CAR T cell-treated mice demonstrated resistance to challenge with CEA-negative CT26 cells.</p><p><strong>Conclusion: </strong>Treatment with CEA-specific, XCL1-secreting CAR-T cells for CEA-positive tumors promoted the generation of CD8<sup>+</sup> T cells against tumor neoantigens, mediating a long-term antitumor immunity against heterogeneous CRCs.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143005727","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
Lipid metabolic remodeling delays senescence of T cells to potentiate their immunity against solid tumors. 脂质代谢重塑延缓T细胞衰老,增强其对实体瘤的免疫力。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-06 DOI: 10.1136/jitc-2024-010403
Yemin Xu, Li Ding, Mengyue Wu, Xiya Wang, Lu Wang, Zhou Xu, Yinhe Xia, Zhennan Cao, Yanqing Zhang, Ruilong Song, Bin Deng, Gang Chen

Background: Tumor cells can drive the senescence of effector T cells by unbalancing their lipid metabolism, thereby limiting adoptive T cell therapy and contributing to tumor immune evasion. Our objective is to provide a feasible strategy for enhancing T cell treatment efficacy against solid tumors.

Methods: In this study, liposomal arachidonyl trifluoromethyl ketone (ATK) was anchored onto the adoptive T cell surface via bioorthogonal reactions, aiming to specifically inhibit the group IVA cytosolic phospholipase A2α (cPLA2α), a key enzyme facilitating phospholipid metabolism and senescent state of T cells.

Results: The surface engineering exerted rare side effects on the activation and migration of T cells, but local and sustained extravasation of ATK downregulated cPLA2α expression, reprogrammed lipid metabolism, and inhibited lipid droplet accumulation. This endows T cells with delayed senescence and declined apoptosis to maintain their tumor-killing potency. Systemic administration of surface-engineered T cells resulted in superior infiltration in solid tumors and improved antitumor efficacy by enhancing the secretion of cytotoxic molecules, thereby prolonging the survival of mice bearing colorectal carcinoma and melanoma xenografts.

Conclusions: Lipid-metabolically remodeled T cells with delayed senescence increase efficacy in tumor microenvironment, highlighting a novel strategy for solid tumor immunotherapy.

背景:肿瘤细胞可以通过失衡效应T细胞的脂质代谢来驱动效应T细胞衰老,从而限制了过继性T细胞治疗并促进肿瘤免疫逃避。我们的目标是提供一种可行的策略来提高T细胞治疗实体瘤的疗效。方法:本研究通过生物正交反应将脂质体花生四烯酰基三氟甲基酮(ATK)锚定在过代T细胞表面,特异性抑制IVA组胞质磷脂酶A2α (cPLA2α),该酶是促进T细胞磷脂代谢和衰老状态的关键酶。结果:表面工程对T细胞的活化和迁移没有明显的副作用,但ATK的局部和持续外渗可下调cPLA2α的表达,重编程脂质代谢,抑制脂滴积累。这使得T细胞延缓衰老和减少凋亡以维持其肿瘤杀伤能力。系统给药表面工程T细胞,使其在实体瘤中具有优越的浸润能力,并通过增强细胞毒性分子的分泌来提高抗肿瘤效果,从而延长结直肠癌和黑色素瘤移植小鼠的存活时间。结论:脂质代谢重塑的延迟衰老的T细胞在肿瘤微环境中提高了疗效,为实体瘤免疫治疗提供了一种新的策略。
{"title":"Lipid metabolic remodeling delays senescence of T cells to potentiate their immunity against solid tumors.","authors":"Yemin Xu, Li Ding, Mengyue Wu, Xiya Wang, Lu Wang, Zhou Xu, Yinhe Xia, Zhennan Cao, Yanqing Zhang, Ruilong Song, Bin Deng, Gang Chen","doi":"10.1136/jitc-2024-010403","DOIUrl":"10.1136/jitc-2024-010403","url":null,"abstract":"<p><strong>Background: </strong>Tumor cells can drive the senescence of effector T cells by unbalancing their lipid metabolism, thereby limiting adoptive T cell therapy and contributing to tumor immune evasion. Our objective is to provide a feasible strategy for enhancing T cell treatment efficacy against solid tumors.</p><p><strong>Methods: </strong>In this study, liposomal arachidonyl trifluoromethyl ketone (ATK) was anchored onto the adoptive T cell surface via bioorthogonal reactions, aiming to specifically inhibit the group IVA cytosolic phospholipase A<sub>2</sub>α (cPLA<sub>2</sub>α), a key enzyme facilitating phospholipid metabolism and senescent state of T cells.</p><p><strong>Results: </strong>The surface engineering exerted rare side effects on the activation and migration of T cells, but local and sustained extravasation of ATK downregulated cPLA2α expression, reprogrammed lipid metabolism, and inhibited lipid droplet accumulation. This endows T cells with delayed senescence and declined apoptosis to maintain their tumor-killing potency. Systemic administration of surface-engineered T cells resulted in superior infiltration in solid tumors and improved antitumor efficacy by enhancing the secretion of cytotoxic molecules, thereby prolonging the survival of mice bearing colorectal carcinoma and melanoma xenografts.</p><p><strong>Conclusions: </strong>Lipid-metabolically remodeled T cells with delayed senescence increase efficacy in tumor microenvironment, highlighting a novel strategy for solid tumor immunotherapy.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749770/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143005701","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
Nivolumab plasma concentration and clearance associated with overall survival in patients with renal cell carcinoma. Nivolumab血浆浓度和清除率与肾细胞癌患者的总生存期相关。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-06 DOI: 10.1136/jitc-2024-010059
Christophe Maritaz, David Combarel, Cécile Dalban, Louis Blondel, Sophie Broutin, Aurelien Marabelle, Laurence Albiges, Angelo Paci

Background: Nivolumab is an immune checkpoint inhibitor (ICI) that selectively inhibits programmed cell death protein 1 activation, restoring antitumor immunity. ICIs are indicated for various types of advanced solid tumors; however, not all patients benefit from them, and tools that could be used in the clinic to predict response to treatment represent an unmet need. Here we describe the development of a new population pharmacokinetic (PPK) model in patients treated with nivolumab in clinical trials. Applying the model to a patient population with renal cell carcinoma identified nivolumab clearance and plasma concentration as predictors of overall survival (OS).

Methods: A custom liquid chromatography with tandem mass spectrometry method for quantifying nivolumab plasma concentration was developed and validated following the European Medicines Agency guidelines for bioanalytical method validation. The PPK model was developed using data from patients treated in the NIVIPIT (n=38) and NIVOREN (n=137) trials of nivolumab in metastatic melanoma and renal cell carcinoma, respectively. The PPK model was used to determine pharmacokinetic (PK) parameters such as baseline clearance and simulate individual clearance changes over time. The relationship between PK characteristics (including clearance at Cycle 1 (CLC1), plasma concentration at Cycle 3 and clinical outcomes was assessed in 137 patients treated in NIVOREN. Kaplan-Meier methodology was used in time-to-event analyses.

Results: In 137 patients, the median nivolumab CLC1 was 6 mL/hour and the median plasma concentration at Cycle 3 was 48 µg/mL. Median follow-up was 21.0 months (95% CI 20.2 to 22.5 months) with a survival rate at 6 months of 91.2% and 77.9% at 12 months. In univariate analysis, OS was significantly higher in patients with CLC1<6 mL/hour versus ≥6 mL/hour (HR 2.2 (95% CI 1.2 to 4.1), p=0.0146). Shorter OS was observed in patients with plasma concentration at Cycle 3 below the median (48 µg/mL) versus those above the median (HR 0.4 (95% CI 0.2 to 0.8), p=0.0069). Multivariate analysis showed a trend towards lower clearance, but this did not reach statistical significance (p=0.0694).

Conclusions: Results of the study may potentially be used to predict outcomes of nivolumab therapy in patients with renal cell carcinoma. Additional applications may include guiding dose adjustments of nivolumab in those who are less likely to respond to the initial dose.

背景:Nivolumab是一种免疫检查点抑制剂(ICI),选择性抑制程序性细胞死亡蛋白1的激活,恢复抗肿瘤免疫。ICIs适用于各种类型的晚期实体瘤;然而,并不是所有的患者都能从中受益,而且可以用于临床预测治疗反应的工具还没有得到满足。在这里,我们描述了一种新的人群药代动力学(PPK)模型的发展,在临床试验中接受纳武单抗治疗的患者。将该模型应用于肾癌患者群体,确定了纳武单抗清除率和血浆浓度作为总生存期(OS)的预测因子。方法:根据欧洲药品管理局生物分析方法验证指南,开发并验证了用于定量纳武单抗血浆浓度的定制液相色谱串联质谱法。PPK模型是根据NIVIPIT (n=38)和NIVOREN (n=137)试验中nivolumab治疗转移性黑色素瘤和肾细胞癌的患者数据开发的。PPK模型用于确定药代动力学(PK)参数,如基线清除率,并模拟个体清除率随时间的变化。在137例接受NIVOREN治疗的患者中,评估了PK特性(包括第1周期清除率(CLC1)、第3周期血药浓度)与临床结果之间的关系。时间-事件分析采用Kaplan-Meier方法。结果:137例患者中,尼武单抗CLC1的中位浓度为6 mL/h, Cycle 3时的中位血浆浓度为48µg/mL。中位随访为21.0个月(95% CI 20.2 ~ 22.5个月),6个月生存率为91.2%,12个月生存率为77.9%。在单因素分析中,clc1患者的OS明显更高。结论:该研究结果可能用于预测肾细胞癌患者纳武单抗治疗的结果。其他应用可能包括对初始剂量不太可能有反应的患者进行纳武单抗的指导剂量调整。
{"title":"Nivolumab plasma concentration and clearance associated with overall survival in patients with renal cell carcinoma.","authors":"Christophe Maritaz, David Combarel, Cécile Dalban, Louis Blondel, Sophie Broutin, Aurelien Marabelle, Laurence Albiges, Angelo Paci","doi":"10.1136/jitc-2024-010059","DOIUrl":"10.1136/jitc-2024-010059","url":null,"abstract":"<p><strong>Background: </strong>Nivolumab is an immune checkpoint inhibitor (ICI) that selectively inhibits programmed cell death protein 1 activation, restoring antitumor immunity. ICIs are indicated for various types of advanced solid tumors; however, not all patients benefit from them, and tools that could be used in the clinic to predict response to treatment represent an unmet need. Here we describe the development of a new population pharmacokinetic (PPK) model in patients treated with nivolumab in clinical trials. Applying the model to a patient population with renal cell carcinoma identified nivolumab clearance and plasma concentration as predictors of overall survival (OS).</p><p><strong>Methods: </strong>A custom liquid chromatography with tandem mass spectrometry method for quantifying nivolumab plasma concentration was developed and validated following the European Medicines Agency guidelines for bioanalytical method validation. The PPK model was developed using data from patients treated in the NIVIPIT (n=38) and NIVOREN (n=137) trials of nivolumab in metastatic melanoma and renal cell carcinoma, respectively. The PPK model was used to determine pharmacokinetic (PK) parameters such as baseline clearance and simulate individual clearance changes over time. The relationship between PK characteristics (including clearance at Cycle 1 (CLC1), plasma concentration at Cycle 3 and clinical outcomes was assessed in 137 patients treated in NIVOREN. Kaplan-Meier methodology was used in time-to-event analyses.</p><p><strong>Results: </strong>In 137 patients, the median nivolumab CLC1 was 6 mL/hour and the median plasma concentration at Cycle 3 was 48 µg/mL. Median follow-up was 21.0 months (95% CI 20.2 to 22.5 months) with a survival rate at 6 months of 91.2% and 77.9% at 12 months. In univariate analysis, OS was significantly higher in patients with CLC1<6 mL/hour versus ≥6 mL/hour (HR 2.2 (95% CI 1.2 to 4.1), p=0.0146). Shorter OS was observed in patients with plasma concentration at Cycle 3 below the median (48 µg/mL) versus those above the median (HR 0.4 (95% CI 0.2 to 0.8), p=0.0069). Multivariate analysis showed a trend towards lower clearance, but this did not reach statistical significance (p=0.0694).</p><p><strong>Conclusions: </strong>Results of the study may potentially be used to predict outcomes of nivolumab therapy in patients with renal cell carcinoma. Additional applications may include guiding dose adjustments of nivolumab in those who are less likely to respond to the initial dose.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749330/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143005705","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
Methodological and conceptual considerations for examining the α-FAtE scoring in unresectable hepatocellular carcinoma.
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-06 DOI: 10.1136/jitc-2024-010840
Güner Akgüner
{"title":"Methodological and conceptual considerations for examining the α-FAtE scoring in unresectable hepatocellular carcinoma.","authors":"Güner Akgüner","doi":"10.1136/jitc-2024-010840","DOIUrl":"https://doi.org/10.1136/jitc-2024-010840","url":null,"abstract":"","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749869/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143023460","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
SIGLEC11 promotes M2 macrophage polarization through AKT-mTOR signaling and facilitates the progression of gastric cancer. SIGLEC11通过AKT-mTOR信号通路促进M2巨噬细胞极化,促进胃癌进展。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-04 DOI: 10.1136/jitc-2024-010162
Jingxin Yin, Yang Lu, Yihao Liu, Qimeng Shi, Minmin Shi, Zhenggang Zhu, Da Fu, Zhenqiang Wang, Chen Li

Background: Sialic acid-binding immunoglobulin-like lectins (SIGLECs) are widely expressed on immune cell surfaces, play an important role in maintaining immune homeostasis and regulating inflammatory responses, and are increasingly emerging as potential targets for tumor immunotherapy. However, the expression profile and crucial role of SIGLEC11 in gastric cancer (GC) remain unclear. This study aimed to elucidate the prognostic relevance of SIGLEC11 expression and its role in the immune microenvironment in patients with GC.

Methods: SIGLEC11 expression profile was analyzed using bioinformatics, immunohistochemistry, and immunofluorescence staining. Flow cytometry, mouse tumor models, patient-derived tumor organoid models, and RNA sequencing were used to explore the potential functions with the underlying mechanisms of SIGLEC11 in a coculture system of macrophages and GC cells.

Results: We demonstrated that SIGLEC11 was predominantly expressed in normal tissues. However, tumor-infiltrating SIGLEC11+ cells in the high SIGLEC11 expression subgroups showed poor overall survival, which was associated with the expression of an immunosuppressive regulator. Our results showed that SIGLEC11 was predominantly expressed in monocytes and macrophages and selectively upregulated in tumor-associated macrophages. Furthermore, SIGLEC11 promoted macrophage M2 polarization via AKT-mTOR signaling. In addition, SIGLEC11+ macrophages accelerate GC progression.

Conclusions: The abundance of SIGLEC11+ M2-like macrophage-infiltrating tumors may serve as a biomarker for identifying immunosuppressive subtypes of GC. Thus, the potential role of SIGLEC11+ M2 macrophages as therapeutic targets warrants further investigation.

背景:唾液酸结合免疫球蛋白样凝集素(SIGLECs)广泛表达于免疫细胞表面,在维持免疫稳态和调节炎症反应中发挥重要作用,并日益成为肿瘤免疫治疗的潜在靶点。然而,SIGLEC11在胃癌(GC)中的表达谱和关键作用尚不清楚。本研究旨在阐明SIGLEC11表达与胃癌患者预后的相关性及其在免疫微环境中的作用。方法:采用生物信息学、免疫组织化学和免疫荧光染色分析SIGLEC11表达谱。通过流式细胞术、小鼠肿瘤模型、患者源性肿瘤类器官模型和RNA测序,研究了SIGLEC11在巨噬细胞和GC细胞共培养系统中的潜在功能及其潜在机制。结果:我们发现SIGLEC11在正常组织中主要表达。然而,在SIGLEC11高表达亚组中,肿瘤浸润的SIGLEC11+细胞表现出较差的总生存率,这与免疫抑制调节因子的表达有关。我们的研究结果表明,SIGLEC11主要在单核细胞和巨噬细胞中表达,并在肿瘤相关巨噬细胞中选择性上调。此外,SIGLEC11通过AKT-mTOR信号通路促进巨噬细胞M2极化。此外,SIGLEC11+巨噬细胞加速GC的进展。结论:SIGLEC11+ m2样巨噬细胞浸润肿瘤的丰度可作为鉴别GC免疫抑制亚型的生物标志物。因此,SIGLEC11+ M2巨噬细胞作为治疗靶点的潜在作用值得进一步研究。
{"title":"SIGLEC11 promotes M2 macrophage polarization through AKT-mTOR signaling and facilitates the progression of gastric cancer.","authors":"Jingxin Yin, Yang Lu, Yihao Liu, Qimeng Shi, Minmin Shi, Zhenggang Zhu, Da Fu, Zhenqiang Wang, Chen Li","doi":"10.1136/jitc-2024-010162","DOIUrl":"10.1136/jitc-2024-010162","url":null,"abstract":"<p><strong>Background: </strong>Sialic acid-binding immunoglobulin-like lectins (SIGLECs) are widely expressed on immune cell surfaces, play an important role in maintaining immune homeostasis and regulating inflammatory responses, and are increasingly emerging as potential targets for tumor immunotherapy. However, the expression profile and crucial role of SIGLEC11 in gastric cancer (GC) remain unclear. This study aimed to elucidate the prognostic relevance of SIGLEC11 expression and its role in the immune microenvironment in patients with GC.</p><p><strong>Methods: </strong>SIGLEC11 expression profile was analyzed using bioinformatics, immunohistochemistry, and immunofluorescence staining. Flow cytometry, mouse tumor models, patient-derived tumor organoid models, and RNA sequencing were used to explore the potential functions with the underlying mechanisms of SIGLEC11 in a coculture system of macrophages and GC cells.</p><p><strong>Results: </strong>We demonstrated that SIGLEC11 was predominantly expressed in normal tissues. However, tumor-infiltrating SIGLEC11<sup>+</sup> cells in the high SIGLEC11 expression subgroups showed poor overall survival, which was associated with the expression of an immunosuppressive regulator. Our results showed that SIGLEC11 was predominantly expressed in monocytes and macrophages and selectively upregulated in tumor-associated macrophages. Furthermore, SIGLEC11 promoted macrophage M2 polarization via AKT-mTOR signaling. In addition, SIGLEC11<sup>+</sup> macrophages accelerate GC progression.</p><p><strong>Conclusions: </strong>The abundance of SIGLEC11<sup>+</sup> M2-like macrophage-infiltrating tumors may serve as a biomarker for identifying immunosuppressive subtypes of GC. Thus, the potential role of SIGLEC11<sup>+</sup> M2 macrophages as therapeutic targets warrants further investigation.</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/PMC11748936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927110","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
Early treatment discontinuation in patients with deficient mismatch repair or microsatellite instability high metastatic colorectal cancer receiving immune checkpoint inhibitors. 错配修复缺陷或微卫星不稳定的高转移性结直肠癌患者接受免疫检查点抑制剂的早期停药
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-04 DOI: 10.1136/jitc-2024-010424
Julien Taieb, Margherita Ambrosini, Emily Alouani, Sara Lonardi, Frank A Sinicrope, Marie Decraecker, Alice Boileve, Emilie Hafliger, Thibault Mazard, Simon Pernot, Pauline Parent, Javier Ros, Michael J Overman, Priya Jayachandran, Vincenzo Nasca, Lisa Salvatore, Rosine Guimbaud, Chiara Cremolini, David Tougeron, Filippo Pietrantonio

Background: Immune checkpoint inhibitors (ICIs) are recommended to treat patients with deficient mismatch repair/microsatellite instability high (dMMR/MSI-H) metastatic colorectal cancer (mCRC). Pivotal trials have fixed a maximum ICI duration of 2 years, without a compelling rationale. A shorter treatment duration has the potential to improve patients' quality of life and reduce both toxicity and cost without compromising efficacy. Here we examine whether early treatment discontinuation (ETD) before 13 months in patients without progressive disease (PD) can lead to similar long-term disease control compared with a longer treatment duration (LTD).

Methods: To assess whether ETD is associated with similar outcomes compared with LTD, we assembled an international cohort of patients with dMMR/MSI-H mCRC treated with ICIs who stopped treatment for a reason other than PD within 395 days (ETD group) and compared them to those who continued for >395 days (LTD group). Outcomes were adjusted for patient/tumor characteristics. Primary endpoint was progression-free survival (PFS) and secondary endpoints were objective response rate (ORR), overall survival (OS) and safety.

Results: Of 976 patients, 137 and 394 were allocated to the ETD and LTD groups, respectively. In the ETD group, treatment was discontinued due to toxicity (n=56), objective response (n=43), surgery (n=28), patient decision (n=2) or other reasons (n=8). Baseline characteristics were well balanced between the two groups: 22% in both groups received both anti-programmed death-(ligand) 1 (anti-PD-(L)1) + anti-cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4); all others received anti-PD-(L)1 monotherapy. ORR to ICIs was 81% in both groups. Median duration of treatment was ~7 months in the ETD and ~24 months in the LTD group. After a median follow-up of 44 months (IQR: 30-67), similar PFS (HR: 0.92, 95% CI: 0.60 to 1.40, p=0.69) and OS (HR: 1.15, 95% CI: 0.66 to 1.99, p=0.62) from the start of ICIs were observed in ETD and LTD patients. In the ETD group, 28 (20%) patients had a PFS event and 9 restarted ICIs with a disease control rate of 66%.

Conclusions: In our international series of dMMR/MSI-H mCRC, ETD of ICIs in the absence of PD did not seem detrimental in terms of PFS and OS compared with continuing treatment beyond 1 year. Randomized clinical trials to compare short and long treatment duration are now warranted.

背景:免疫检查点抑制剂(ICIs)被推荐用于治疗缺陷错配修复/微卫星不稳定性高(dMMR/MSI-H)转移性结直肠癌(mCRC)患者。关键试验已确定最长ICI持续时间为2年,但没有令人信服的理由。较短的治疗时间有可能改善患者的生活质量,在不影响疗效的情况下降低毒性和成本。在这里,我们研究了与较长的治疗时间(LTD)相比,未进展性疾病(PD)患者在13个月前早期停止治疗(ETD)是否可以导致类似的长期疾病控制。方法:为了评估ETD与LTD相比是否与类似的结果相关,我们收集了一个国际队列,包括接受ICIs治疗的dMMR/MSI-H mCRC患者,这些患者在395天内因PD以外的原因停止治疗(ETD组),并将其与持续治疗395天(LTD组)的患者进行比较。结果根据患者/肿瘤特征进行调整。主要终点是无进展生存期(PFS),次要终点是客观缓解率(ORR)、总生存期(OS)和安全性。结果:976例患者中,ETD组137例,LTD组394例。在ETD组中,由于毒性(n=56)、客观反应(n=43)、手术(n=28)、患者决定(n=2)或其他原因(n=8)而停止治疗。两组之间的基线特征很好地平衡:两组中22%同时接受抗程序性死亡-(配体)1(抗pd -(L)1) +抗细胞毒性t淋巴细胞抗原-4(抗ctla -4);其余均接受抗pd -(L)1单药治疗。两组对ICIs的ORR均为81%。ETD组和LTD组的中位治疗时间分别为~7个月和~24个月。中位随访44个月(IQR: 30-67)后,ETD和LTD患者从开始使用ici开始观察到相似的PFS (HR: 0.92, 95% CI: 0.60 ~ 1.40, p=0.69)和OS (HR: 1.15, 95% CI: 0.66 ~ 1.99, p=0.62)。在ETD组中,28例(20%)患者出现PFS事件,9例重新启动ICIs,疾病控制率为66%。结论:在我们的dMMR/MSI-H mCRC国际系列中,与持续治疗1年以上相比,ICIs在无PD的情况下的ETD似乎对PFS和OS没有不利影响。现在有必要进行随机临床试验来比较短期和长期治疗时间。
{"title":"Early treatment discontinuation in patients with deficient mismatch repair or microsatellite instability high metastatic colorectal cancer receiving immune checkpoint inhibitors.","authors":"Julien Taieb, Margherita Ambrosini, Emily Alouani, Sara Lonardi, Frank A Sinicrope, Marie Decraecker, Alice Boileve, Emilie Hafliger, Thibault Mazard, Simon Pernot, Pauline Parent, Javier Ros, Michael J Overman, Priya Jayachandran, Vincenzo Nasca, Lisa Salvatore, Rosine Guimbaud, Chiara Cremolini, David Tougeron, Filippo Pietrantonio","doi":"10.1136/jitc-2024-010424","DOIUrl":"10.1136/jitc-2024-010424","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) are recommended to treat patients with deficient mismatch repair/microsatellite instability high (dMMR/MSI-H) metastatic colorectal cancer (mCRC). Pivotal trials have fixed a maximum ICI duration of 2 years, without a compelling rationale. A shorter treatment duration has the potential to improve patients' quality of life and reduce both toxicity and cost without compromising efficacy. Here we examine whether early treatment discontinuation (ETD) before 13 months in patients without progressive disease (PD) can lead to similar long-term disease control compared with a longer treatment duration (LTD).</p><p><strong>Methods: </strong>To assess whether ETD is associated with similar outcomes compared with LTD, we assembled an international cohort of patients with dMMR/MSI-H mCRC treated with ICIs who stopped treatment for a reason other than PD within 395 days (ETD group) and compared them to those who continued for >395 days (LTD group). Outcomes were adjusted for patient/tumor characteristics. Primary endpoint was progression-free survival (PFS) and secondary endpoints were objective response rate (ORR), overall survival (OS) and safety.</p><p><strong>Results: </strong>Of 976 patients, 137 and 394 were allocated to the ETD and LTD groups, respectively. In the ETD group, treatment was discontinued due to toxicity (n=56), objective response (n=43), surgery (n=28), patient decision (n=2) or other reasons (n=8). Baseline characteristics were well balanced between the two groups: 22% in both groups received both anti-programmed death-(ligand) 1 (anti-PD-(L)1) + anti-cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4); all others received anti-PD-(L)1 monotherapy. ORR to ICIs was 81% in both groups. Median duration of treatment was ~7 months in the ETD and ~24 months in the LTD group. After a median follow-up of 44 months (IQR: 30-67), similar PFS (HR: 0.92, 95% CI: 0.60 to 1.40, p=0.69) and OS (HR: 1.15, 95% CI: 0.66 to 1.99, p=0.62) from the start of ICIs were observed in ETD and LTD patients. In the ETD group, 28 (20%) patients had a PFS event and 9 restarted ICIs with a disease control rate of 66%.</p><p><strong>Conclusions: </strong>In our international series of dMMR/MSI-H mCRC, ETD of ICIs in the absence of PD did not seem detrimental in terms of PFS and OS compared with continuing treatment beyond 1 year. Randomized clinical trials to compare short and long treatment duration are now warranted.</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/PMC11749684/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927105","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
Effect of radiotherapy exposure on fruquintinib plus sintilimab treatment in refractory microsatellite stable metastatic colorectal cancer: a prospective observation study. 放疗暴露对氟喹替尼联合辛替单抗治疗难治性微卫星稳定转移性结直肠癌的影响:一项前瞻性观察研究。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-04 DOI: 10.1136/jitc-2024-009415
Mingxia Cheng, Min Jin, Shengli Yang, Lei Zhao, Dandan Yu, Zhenyu Lin, Pindong Li, Chuying Huang, Junli Liu, Jing Wang, Jun Xue, Hong Ma, Jianli Hu, Kunyu Yang, Tao Zhang, Hongli Liu

Background: Immune checkpoint inhibitors (ICIs) in combination with antiangiogenic drugs have shown promising outcomes in the third-line and subsequent treatments of patients with microsatellite stable metastatic colorectal cancer (MSS-mCRC). Radiotherapy (RT) may enhance the antitumor effect of immunotherapy. However, the effect of RT exposure on patients receiving ICIs and targeted therapy remains unclear. This study aimed to investigate the association between RT exposure and clinical responses to fruquintinib (a highly selective tyrosine kinase inhibitor of vascular endothelial growth factor receptor) plus sintilimab (an anti-programmed death 1 antibody; F&S) in previously treated patients with MSS-mCRC and to explore predictive biomarkers.

Methods: In this prospective observational study, patients with mCRC receiving F&S as third-line or subsequent treatment were enrolled. Eligible patients were divided into the RT cohort (RTC) and the non-RT cohort (NRTC) according to their RT history. The primary endpoint was the objective response rate (ORR). Secondary endpoints included disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety. Pretreatment fecal and serum samples were collected for microbiome analysis, metabolome analysis, and immune signatures to identify biomarkers for treatment.

Results: A total of 55 patients were included, of which 25 were in the RTC and 30 in the NRTC. Better ORR (28.0% vs 6.7%, p=0.048), DCR (80.0% vs 36.7%, p=0.002), median PFS (6.2 vs 2.7 months, p<0.001), and median OS (14.8 vs 5.9 months, p=0.019) were noted in patients with RTC than those with NRTC. The enrichment of Lactobacillus, Bifidobacterium, and PC(20:5(5Z,8Z,11Z,14Z,17Z)/20:3(8Z,11Z,14Z)) in RTC significantly predicted better DCR and PFS, whereas guanosine and interleukin-10 predominated in patients with NRTC were negatively correlated with PFS and OS.

Conclusions: Patients with RT exposure benefited significantly from F&S in the third-line or subsequent treatment for MSS-mCRC. Gut microbiota, metabolites, and cytokines may help predict F&S outcomes for mCRC, which may be helpful in treatment decision-making.

Trial registration number: ClinicalTrials.gov identifier: NCT05635149.

背景:免疫检查点抑制剂(ICIs)联合抗血管生成药物在微卫星稳定转移性结直肠癌(MSS-mCRC)患者的三线和后续治疗中显示出良好的结果。放射治疗可以增强免疫治疗的抗肿瘤作用。然而,RT暴露对接受ICIs和靶向治疗的患者的影响尚不清楚。本研究旨在探讨RT暴露与fruquininib(一种血管内皮生长因子受体的高选择性酪氨酸激酶抑制剂)和sintilimab(一种抗程序性死亡1抗体;F&S)在既往治疗过的MSS-mCRC患者中,并探索预测性生物标志物。方法:在这项前瞻性观察研究中,纳入了接受F&S作为三线或后续治疗的mCRC患者。将符合条件的患者根据其RT病史分为RT组(RTC)和非RT组(NRTC)。主要终点为客观缓解率(ORR)。次要终点包括疾病控制率(DCR)、无进展生存期(PFS)、总生存期(OS)和安全性。收集预处理粪便和血清样本进行微生物组分析、代谢组分析和免疫标记,以确定治疗的生物标志物。结果:共纳入55例患者,其中RTC组25例,NRTC组30例。RTC患者较好的ORR (28.0% vs 6.7%, p=0.048)、DCR (80.0% vs 36.7%, p=0.002)、中位PFS (6.2 vs 2.7个月)、pLactobacillus、Bifidobacterium和PC(20:5(5Z,8Z,11Z,14Z,17Z)/20:3(8Z,11Z,14Z)显著预测较好的DCR和PFS,而NRTC患者以鸟苷和白介素-10为主与PFS和OS呈负相关。结论:放疗暴露患者在MSS-mCRC的三线或后续治疗中显著受益于F&S。肠道菌群、代谢物和细胞因子可能有助于预测mCRC的F&S结果,这可能有助于治疗决策。试验注册号:ClinicalTrials.gov标识符:NCT05635149。
{"title":"Effect of radiotherapy exposure on fruquintinib plus sintilimab treatment in refractory microsatellite stable metastatic colorectal cancer: a prospective observation study.","authors":"Mingxia Cheng, Min Jin, Shengli Yang, Lei Zhao, Dandan Yu, Zhenyu Lin, Pindong Li, Chuying Huang, Junli Liu, Jing Wang, Jun Xue, Hong Ma, Jianli Hu, Kunyu Yang, Tao Zhang, Hongli Liu","doi":"10.1136/jitc-2024-009415","DOIUrl":"https://doi.org/10.1136/jitc-2024-009415","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) in combination with antiangiogenic drugs have shown promising outcomes in the third-line and subsequent treatments of patients with microsatellite stable metastatic colorectal cancer (MSS-mCRC). Radiotherapy (RT) may enhance the antitumor effect of immunotherapy. However, the effect of RT exposure on patients receiving ICIs and targeted therapy remains unclear. This study aimed to investigate the association between RT exposure and clinical responses to fruquintinib (a highly selective tyrosine kinase inhibitor of vascular endothelial growth factor receptor) plus sintilimab (an anti-programmed death 1 antibody; F&S) in previously treated patients with MSS-mCRC and to explore predictive biomarkers.</p><p><strong>Methods: </strong>In this prospective observational study, patients with mCRC receiving F&S as third-line or subsequent treatment were enrolled. Eligible patients were divided into the RT cohort (RTC) and the non-RT cohort (NRTC) according to their RT history. The primary endpoint was the objective response rate (ORR). Secondary endpoints included disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety. Pretreatment fecal and serum samples were collected for microbiome analysis, metabolome analysis, and immune signatures to identify biomarkers for treatment.</p><p><strong>Results: </strong>A total of 55 patients were included, of which 25 were in the RTC and 30 in the NRTC. Better ORR (28.0% vs 6.7%, p=0.048), DCR (80.0% vs 36.7%, p=0.002), median PFS (6.2 vs 2.7 months, p<0.001), and median OS (14.8 vs 5.9 months, p=0.019) were noted in patients with RTC than those with NRTC. The enrichment of <i>Lactobacillus</i>, <i>Bifidobacterium</i>, and PC(20:5(5Z,8Z,11Z,14Z,17Z)/20:3(8Z,11Z,14Z)) in RTC significantly predicted better DCR and PFS, whereas guanosine and interleukin-10 predominated in patients with NRTC were negatively correlated with PFS and OS.</p><p><strong>Conclusions: </strong>Patients with RT exposure benefited significantly from F&S in the third-line or subsequent treatment for MSS-mCRC. Gut microbiota, metabolites, and cytokines may help predict F&S outcomes for mCRC, which may be helpful in treatment decision-making.</p><p><strong>Trial registration number: </strong>ClinicalTrials.gov identifier: NCT05635149.</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":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927107","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
Depletion of myeloid-derived suppressor cells sensitizes murine multiple myeloma to PD-1 checkpoint inhibitors. 骨髓源性抑制细胞的耗竭使小鼠多发性骨髓瘤对PD-1检查点抑制剂敏感。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-04 DOI: 10.1136/jitc-2024-008979
Wei Xiong, Liuling Xiao, Rui Duan, Qiang Wang, Miao Xian, Chuanchao Zhang, Pan Su, Yabo Li, Ling Zhong, Jianfei Qian, Chengyun Zheng, Qing Yi

Background: Cancer immunotherapy using immune checkpoint blockade (ICB) has revolutionized cancer treatment. However, patients with multiple myeloma (MM) rarely respond to ICB. Accumulating evidence indicates that the complicated tumor microenvironment (TME) significantly impacts the efficacy of ICB therapy. Therefore, investigating how TME components in MM influence ICB treatment is urgent.

Methods: We employed two well-established murine myeloma models, 5TGM1 and Vk*MYC, by intravenously injecting 5TGM1 or Vk*MYC cells into mice, respectively, to determine ICB therapeutic efficacy in MM. Total mouse IgG or Ig2b ELISA or QuickGel split beta SPE kits and in vivo bioluminescent imaging were used to monitor MM tumor burden. Cytometry by time of flight (CyTOF) was used to quantify MM TME components. T cell proliferation and function were detected using flow cytometry. Peptide-Fc fusion proteins were used to deplete myeloid-derived suppressor cells (MDSCs). MMDTR, Foxp3DTR, CD4 KO and CD8 KO mice were used to elucidate the underlying mechanisms. Gene expression levels in human MM were analyzed using Gene Expression Omnibus public datasets.

Results: We found that programmed cell death protein 1 (PD-1) antibody treatment had a therapeutic effect in 5TGM1 mice; it was ineffective in Vk*MYC mice. CyTOF indicated that the bone marrow (BM) of both models was inflamed, suggesting that immune suppressive cells might be inhibiting the reactivation of T cells in the BM. We observed higher numbers of MDSCs, regulatory T (Treg) cells, and tumor-associated macrophage (TAMs) in myeloma BM compared with that of tumor-free mice. Specifically, depleting MDSCs, but not Treg cells or TAMs, sensitized Vk*MYC mice and enhanced the response of 5TGM1 mice to PD-1 ICB, which was dependent on CD8+ but not CD4+ T cells. MDSCs, especially M-MDSCs and CD84+ MDSCs, significantly inhibited the activation and cytotoxic cytokine production of CD8+ T cells in vitro. Moreover, database profiling of patient BM revealed a negative correlation between MDSCs signature genes and cytotoxic CD8+ T cell signature genes, with post-maintenance patients with myeloma displaying a higher ratio of cytotoxic CD8+ T cell to MDSCs signature genes compared with pretreated patients.

Conclusion: Our study highlights the potential of MDSCs depletion in enhancing the sensitivity of patients with myeloma to PD-1 ICB therapy.

背景:使用免疫检查点阻断(ICB)的癌症免疫治疗已经彻底改变了癌症治疗。然而,多发性骨髓瘤(MM)患者很少对ICB有反应。越来越多的证据表明,复杂的肿瘤微环境(TME)显著影响ICB治疗的疗效。因此,研究MM中TME成分如何影响ICB治疗是迫在眉睫的。方法:采用两种已建立的小鼠骨髓瘤模型5TGM1和Vk*MYC,分别静脉注射5TGM1或Vk*MYC细胞,检测ICB对MM的治疗效果,采用小鼠总IgG或Ig2b ELISA或QuickGel分裂β SPE试剂盒和体内生物荧光成像监测MM肿瘤负荷。用飞行时间细胞术(CyTOF)定量MM TME成分。流式细胞术检测T细胞增殖及功能。使用肽fc融合蛋白来消耗髓源性抑制细胞(MDSCs)。使用MMDTR、Foxp3DTR、CD4 KO和CD8 KO小鼠来阐明其潜在机制。使用Gene expression Omnibus公共数据集分析人MM的基因表达水平。结果:我们发现程序性细胞死亡蛋白1 (PD-1)抗体治疗对5TGM1小鼠有治疗作用;对Vk*MYC小鼠无效。CyTOF显示两种模型的骨髓(BM)都有炎症,提示免疫抑制细胞可能抑制了BM中T细胞的再激活。我们观察到与无瘤小鼠相比,骨髓瘤BM中MDSCs、调节性T (Treg)细胞和肿瘤相关巨噬细胞(tam)的数量更高。具体来说,消耗MDSCs,而不是Treg细胞或tam,使Vk*MYC小鼠增敏,并增强5TGM1小鼠对PD-1 ICB的反应,PD-1 ICB依赖于CD8+而不是CD4+ T细胞。MDSCs,特别是M-MDSCs和CD84+ MDSCs,在体外显著抑制CD8+ T细胞的活化和细胞毒细胞因子的产生。此外,患者BM的数据库分析显示MDSCs特征基因与细胞毒性CD8+ T细胞特征基因之间存在负相关,与治疗前的患者相比,维持后的骨髓瘤患者显示出更高的细胞毒性CD8+ T细胞与MDSCs特征基因的比例。结论:我们的研究强调了MDSCs消耗在增强骨髓瘤患者对PD-1 ICB治疗的敏感性方面的潜力。
{"title":"Depletion of myeloid-derived suppressor cells sensitizes murine multiple myeloma to PD-1 checkpoint inhibitors.","authors":"Wei Xiong, Liuling Xiao, Rui Duan, Qiang Wang, Miao Xian, Chuanchao Zhang, Pan Su, Yabo Li, Ling Zhong, Jianfei Qian, Chengyun Zheng, Qing Yi","doi":"10.1136/jitc-2024-008979","DOIUrl":"10.1136/jitc-2024-008979","url":null,"abstract":"<p><strong>Background: </strong>Cancer immunotherapy using immune checkpoint blockade (ICB) has revolutionized cancer treatment. However, patients with multiple myeloma (MM) rarely respond to ICB. Accumulating evidence indicates that the complicated tumor microenvironment (TME) significantly impacts the efficacy of ICB therapy. Therefore, investigating how TME components in MM influence ICB treatment is urgent.</p><p><strong>Methods: </strong>We employed two well-established murine myeloma models, 5TGM1 and Vk*MYC, by intravenously injecting 5TGM1 or Vk*MYC cells into mice, respectively, to determine ICB therapeutic efficacy in MM. Total mouse IgG or Ig2b ELISA or QuickGel split beta SPE kits and in vivo bioluminescent imaging were used to monitor MM tumor burden. Cytometry by time of flight (CyTOF) was used to quantify MM TME components. T cell proliferation and function were detected using flow cytometry. Peptide-Fc fusion proteins were used to deplete myeloid-derived suppressor cells (MDSCs). MM<sup>DTR</sup>, Foxp3<sup>DTR</sup>, CD4 KO and CD8 KO mice were used to elucidate the underlying mechanisms. Gene expression levels in human MM were analyzed using Gene Expression Omnibus public datasets.</p><p><strong>Results: </strong>We found that programmed cell death protein 1 (PD-1) antibody treatment had a therapeutic effect in 5TGM1 mice; it was ineffective in Vk*MYC mice. CyTOF indicated that the bone marrow (BM) of both models was inflamed, suggesting that immune suppressive cells might be inhibiting the reactivation of T cells in the BM. We observed higher numbers of MDSCs, regulatory T (Treg) cells, and tumor-associated macrophage (TAMs) in myeloma BM compared with that of tumor-free mice. Specifically, depleting MDSCs, but not Treg cells or TAMs, sensitized Vk*MYC mice and enhanced the response of 5TGM1 mice to PD-1 ICB, which was dependent on CD8<sup>+</sup> but not CD4<sup>+</sup> T cells. MDSCs, especially M-MDSCs and CD84<sup>+</sup> MDSCs, significantly inhibited the activation and cytotoxic cytokine production of CD8<sup>+</sup> T cells in vitro. Moreover, database profiling of patient BM revealed a negative correlation between MDSCs signature genes and cytotoxic CD8<sup>+</sup> T cell signature genes, with post-maintenance patients with myeloma displaying a higher ratio of cytotoxic CD8<sup>+</sup> T cell to MDSCs signature genes compared with pretreated patients.</p><p><strong>Conclusion: </strong>Our study highlights the potential of MDSCs depletion in enhancing the sensitivity of patients with myeloma to PD-1 ICB therapy.</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/PMC11749690/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting Siglec-E facilitates tumor vaccine-induced antitumor immunity in renal carcinoma. 靶向siglece促进肿瘤疫苗诱导的肾癌抗肿瘤免疫。
IF 10.3 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-04 DOI: 10.1136/jitc-2024-010521
Yanyan Zheng, Jiawei Wang, Guangya Zhao, Zichun Zhang, Yingxiang Shao, Bowen Lu, Yuchen Zhang, Renjin Chen, Li Sun, Xiaohui Xie, Jiage Ding, Junnian Zheng, Dafei Chai

Background: Siglec-E is an immune checkpoint inhibitory molecule. Expression of Siglec-E on the immune cells has been shown to promote tumor regression. This study aimed to develop an adenovirus (Ad) vaccine targeting Siglec-E and carbonic anhydrase IX (CAIX) (Ad-Siglec-E/CAIX) and to evaluate its potential antitumor effects in several preclinical renal cancer models.

Methods: Ad vaccines encoding Siglec-E or CAIX were developed and evaluated for their therapeutic potential in mouse subcutaneous, lung metastatic, and orthotopic tumor models. The expression of Ad-Siglec-E/CAIX was confirmed via PCR and flow cytometry. Immune responses induced by Ad-Siglec-E/CAIX were assessed in vitro and in vivo using flow cytometry, immunohistochemistry, ELISA, histological analysis, cell proliferation, enzyme-linked immunosorbent spot, cytotoxic T lymphocytes (CTL) killing, and cell depletion assays.

Results: Ad-Siglec-E/CAIX vaccine induced the increase of tumor-infiltrated immune cells, and significantly suppressed the subcutaneous tumor growth of renal carcinoma. Immunization with Ad-Siglec-E/CAIX promoted the induction and maturation of CD11c+ dendritic cells and their subsets, which in turn enhanced tumor-specific CD8+ T cell immune responses, as evidenced by increased CD8+ T cell proliferation and CTL activity. Importantly, the deletion of CD8+ T cells in vivo abolished the antitumor effect of the Ad-Siglec-E/CAIX vaccine, highlighting the essential role of functional CD8+ T cell responses. The potent therapeutic efficacy of the Ad-Siglec-E/CAIX vaccine was also observed in lung metastasis and orthotopic tumor models through tumor-specific CD8+ T cell immune responses.

Conclusions: Our results indicate that targeting Siglec-E enhances the therapeutic efficacy of Ad-CAIX against renal carcinoma, providing a promising therapeutic option for solid tumors.

背景:siglece是一种免疫检查点抑制分子。免疫细胞上siglece的表达已被证明可促进肿瘤消退。本研究旨在开发一种针对siglece和碳酸酐酶IX (CAIX) (Ad- siglece /CAIX)的腺病毒(Ad)疫苗,并在几种临床前肾癌模型中评估其潜在的抗肿瘤作用。方法:开发编码siglece或CAIX的Ad疫苗,并评估其在小鼠皮下、肺转移和原位肿瘤模型中的治疗潜力。通过PCR和流式细胞术证实ad - siglece /CAIX的表达。通过流式细胞术、免疫组织化学、ELISA、组织学分析、细胞增殖、酶联免疫吸附斑点、细胞毒性T淋巴细胞(CTL)杀伤和细胞耗损试验,在体外和体内评估ad - siglece - e /CAIX诱导的免疫应答。结果:ad - siglece /CAIX疫苗诱导肿瘤浸润免疫细胞增加,显著抑制肾癌皮下肿瘤生长。ad - siglece /CAIX免疫促进CD11c+树突状细胞及其亚群的诱导和成熟,进而增强肿瘤特异性CD8+ T细胞免疫应答,CD8+ T细胞增殖和CTL活性增加。重要的是,体内CD8+ T细胞的缺失消除了ad - siglece - e /CAIX疫苗的抗肿瘤作用,突出了功能性CD8+ T细胞反应的重要作用。通过肿瘤特异性CD8+ T细胞免疫应答,还观察到ad - siglece /CAIX疫苗在肺转移和原位肿瘤模型中的有效治疗效果。结论:我们的研究结果表明,靶向siglece可增强Ad-CAIX对肾癌的治疗效果,为实体瘤的治疗提供了一个有希望的选择。
{"title":"Targeting Siglec-E facilitates tumor vaccine-induced antitumor immunity in renal carcinoma.","authors":"Yanyan Zheng, Jiawei Wang, Guangya Zhao, Zichun Zhang, Yingxiang Shao, Bowen Lu, Yuchen Zhang, Renjin Chen, Li Sun, Xiaohui Xie, Jiage Ding, Junnian Zheng, Dafei Chai","doi":"10.1136/jitc-2024-010521","DOIUrl":"10.1136/jitc-2024-010521","url":null,"abstract":"<p><strong>Background: </strong>Siglec-E is an immune checkpoint inhibitory molecule. Expression of Siglec-E on the immune cells has been shown to promote tumor regression. This study aimed to develop an adenovirus (Ad) vaccine targeting Siglec-E and carbonic anhydrase IX (CAIX) (Ad-Siglec-E/CAIX) and to evaluate its potential antitumor effects in several preclinical renal cancer models.</p><p><strong>Methods: </strong>Ad vaccines encoding Siglec-E or CAIX were developed and evaluated for their therapeutic potential in mouse subcutaneous, lung metastatic, and orthotopic tumor models. The expression of Ad-Siglec-E/CAIX was confirmed via PCR and flow cytometry. Immune responses induced by Ad-Siglec-E/CAIX were assessed in vitro and in vivo using flow cytometry, immunohistochemistry, ELISA, histological analysis, cell proliferation, enzyme-linked immunosorbent spot, cytotoxic T lymphocytes (CTL) killing, and cell depletion assays.</p><p><strong>Results: </strong>Ad-Siglec-E/CAIX vaccine induced the increase of tumor-infiltrated immune cells, and significantly suppressed the subcutaneous tumor growth of renal carcinoma. Immunization with Ad-Siglec-E/CAIX promoted the induction and maturation of CD11c<sup>+</sup> dendritic cells and their subsets, which in turn enhanced tumor-specific CD8<sup>+</sup> T cell immune responses, as evidenced by increased CD8<sup>+</sup> T cell proliferation and CTL activity. Importantly, the deletion of CD8<sup>+</sup> T cells in vivo abolished the antitumor effect of the Ad-Siglec-E/CAIX vaccine, highlighting the essential role of functional CD8<sup>+</sup> T cell responses. The potent therapeutic efficacy of the Ad-Siglec-E/CAIX vaccine was also observed in lung metastasis and orthotopic tumor models through tumor-specific CD8<sup>+</sup> T cell immune responses.</p><p><strong>Conclusions: </strong>Our results indicate that targeting Siglec-E enhances the therapeutic efficacy of Ad-CAIX against renal carcinoma, providing a promising therapeutic option for solid tumors.</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/PMC11749828/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142927114","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